US11680104B2 - Anti-LAG-3 antibodies - Google Patents

Anti-LAG-3 antibodies Download PDF

Info

Publication number
US11680104B2
US11680104B2 US15/756,767 US201615756767A US11680104B2 US 11680104 B2 US11680104 B2 US 11680104B2 US 201615756767 A US201615756767 A US 201615756767A US 11680104 B2 US11680104 B2 US 11680104B2
Authority
US
United States
Prior art keywords
seq
amino acid
acid sequence
antibody
lag
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US15/756,767
Other languages
English (en)
Other versions
US20190153112A1 (en
Inventor
Frédéric Triebel
Chrystelle Brignone
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immutep SAS
Original Assignee
Immutep SAS
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB1515572.4A external-priority patent/GB201515572D0/en
Priority claimed from GBGB1612437.2A external-priority patent/GB201612437D0/en
Application filed by Immutep SAS filed Critical Immutep SAS
Assigned to IMMUTEP S.A.S reassignment IMMUTEP S.A.S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRIGNONE, Chrystelle, TRIEBEL, FREDERIC
Publication of US20190153112A1 publication Critical patent/US20190153112A1/en
Assigned to IMMUTEP S.A.S. reassignment IMMUTEP S.A.S. CHANGE OF ADDRESS Assignors: IMMUTEP S.A.S.
Application granted granted Critical
Publication of US11680104B2 publication Critical patent/US11680104B2/en
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2896Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against molecules with a "CD"-designation, not provided for elsewhere
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/51Complete heavy chain or Fd fragment, i.e. VH + CH1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/515Complete light chain, i.e. VL + CL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • This invention relates to antibodies, or antigen-binding fragments thereof, that bind to Lymphocyte-activation gene-3 (LAG-3), especially antibodies, or antigen-binding fragments thereof, that are agonists of LAG-3, and to use of the antibodies or fragments as medicaments, in particular for the treatment of conditions associated with proliferation and/or activation of CD4 + and/or CD8 + T cells, in particular inflammatory and autoimmune disorders.
  • LAG-3 Lymphocyte-activation gene-3
  • the lymphocyte activation gene 3 (LAG-3) is a CD4 homolog type I membrane protein with four extracellular Ig superfamily domains. Similar to CD4, LAG-3 oligomerizes at the surfaces of T cells and binds to MHC class II molecules on antigen-presenting cells (APCs) but with significantly higher affinity than CD4. LAG-3 is expressed on activated CD4-positive and CD8-positive T lymphocytes where it associates with the CD3-TCR complex at the cell surface and negatively regulates signal transduction. As a consequence, it negatively regulates T cell proliferation, function, and homeostasis.
  • APCs antigen-presenting cells
  • LAG-3 also encodes an alternative splice variant that is translated to a soluble form of LAG-3 (sLAG-3).
  • LAG-3 activates antigen-presenting cells (APCs) through MHC class II signalling, leading to increased antigen-specific T-cell responses in vivo (Triebel, Trends Immunol., 2003, 24: 619-622).
  • the amino acid sequence of human and murine LAG-3 protein is provided in Figure 1 of Huard et al ( Proc. Natl. Acad. Sci . USA, 11: 5744-5749, 1997).
  • the sequence of human LAG-3 protein is repeated in FIG. 1 below (SEQ ID NO: 27).
  • the amino acid sequences of the four extracellular Ig superfamily domains (D1, D2, D3, and D4) of human LAG-3 are at amino acid residues: 1-149 (D1) (SEQ ID NO:28); 150-239 (D2) (SEQ ID NO:29); 240-330 (D3) (SEQ ID NO:39); and 331-412 (D4) (SEQ ID NO:51).
  • Baixeras, et al. J. Exp. Med., 1992, Vol. 176: 327-337) describes production of 1764, a mouse monoclonal antibody (isotype IgG1) to human LAG-3 protein. This antibody recognizes a 30 amino acid extra-loop of the first N-terminal D1 domain of human LAG-3. 17B4 inhibits LAG-3/MHC class interactions, and increases T cell proliferation as a LAG-3 signalling antagonist (Huard at al, Eur J Immunol. 1996; 26:1180-6).
  • 17B4 mAb has no agonist activity, as determined by its inability to induce intracellular free calcium elevation into T cells in the absence of a secondary cross-linking reagent (Hannier et al, J Immunol. 1998; 161:4058-65.).
  • Agents capable of modulating the activation and/or effector functions of CD8-positive and CD4-positive T cells are highly desirable.
  • many autoimmune disorders are known to involve autoreactive T cells and autoantibodies.
  • agents that are capable of inhibiting or eliminating autoreactive lymphocytes without compromising the immune system's ability to defend against pathogens.
  • LAG-3 agonists will negatively regulate T cell proliferation and/or function without depleting the T cells, and that such agonists can also be used to treat inflammatory or autoimmune disorders.
  • the applicant has been able to produce monoclonal anti-LAG-3 agonistic antibodies. These antibodies inhibit antigen-induced proliferation of CD4-positive and CD8-positive T cells. Such antibodies, and antigen-binding fragments thereof, can be used for the treatment of immune disorders, in particular T-cell-mediated immune disorders, including inflammatory and autoimmune disorders.
  • an agonistic anti-LAG-3 antibody or an antigen-binding fragment thereof.
  • the antibody is a monoclonal agonistic anti-LAG-3 antibody, or an antigen-binding fragment thereof.
  • LAG-3 refers to Lymphocyte Activation Gene-3.
  • LAG-3 includes variants, isoforms, homologs, orthologs and paralogs.
  • antibodies specific for a human LAG-3 protein may, in certain cases, cross-react with a LAG-3 protein from a species other than human.
  • the antibodies specific for a human LAG-3 protein may be completely specific for the human LAG-3 protein and may not exhibit species or other types of cross-reactivity, or may cross-react with LAG-3 from certain other species but not all other species (e.g., cross-react with monkey LAG-3 but not mouse LAG-3).
  • human LAG-3 refers to human sequence LAG-3, such as the complete amino acid sequence of human LAG-3 having Genbank Accession No. NP 002277 (SEQ ID NO: 38), or the amino acid sequence of human LAG-3 protein given in FIG. 1 (SEQ ID NO: 27).
  • mouse LAG-3 refers to mouse sequence LAG-3, such as the complete amino acid sequence of mouse LAG-3 having Genbank Accession No. NP_032505.
  • LAG-3 is also known in the art as, for example, CD223.
  • the human LAG-3 sequence may differ from human LAG-3 of Genbank Accession No.
  • NP_002277 by having, e.g., conserved mutations or mutations in non-conserved regions and the LAG-3 has substantially the same biological function as the human LAG-3 of Genbank Accession No. NP_002277.
  • a biological function of human LAG-3 is having an epitope in the extracellular domain of LAG-3 that is specifically bound by an antibody of the instant disclosure or a biological function of human LAG-3 is binding to MHC Class II molecules.
  • monkey LAG-3 is intended to encompass LAG-3 proteins expressed by Old World and New World monkeys, including but not limited to cynomolgus monkey LAG-3 and rhesus monkey LAG-3.
  • a representative amino acid sequence for monkey LAG-3 is the rhesus monkey LAG-3 amino acid sequence which is also deposited as Genbank Accession No. XM_001108923.
  • Another representative amino acid sequence for monkey LAG-3 is the alternative rhesus monkey sequence of clone pa23-5 as described in US 2011/0150892 A1. This alternative rhesus sequence exhibits a single amino acid difference, at position 419, as compared to the Genbank-deposited sequence.
  • a particular human LAG-3 sequence will generally be at least 90% identical in amino acid sequence to human LAG-3 of Genbank Accession No. NP_002277 and contains amino acid residues that identify the amino acid sequence as being human when compared to LAG-3 amino acid sequences of other species (e.g., murine).
  • a human LAG-3 can be at least 95%, or even at least 96%, 97%, 98%, or 99% identical in amino acid sequence to LAG-3 of Genbank Accession No. NP_002277.
  • a human LAG-3 sequence will display no more than 10 amino acid differences from the LAG-3 sequence of Genbank Accession No. NP_002277.
  • the human LAG-3 can display no more than 5, or even no more than 4, 3, 2, or 1 amino acid difference from the LAG-3 sequence of Genbank Accession No. NP_002277. Percent identity can be determined as described herein.
  • an agonistic anti-LAG-3 antibody of the invention inhibits antigen-induced CD4 + and/or CD8 + T cell proliferation, or antigen-induced CD4 + and/or CD8 + T cell activation.
  • An agonistic anti-LAG-3 antibody of the invention, or antigen-binding fragment thereof may be an isolated agonistic anti-LAG-3 antibody, or antigen-binding fragment thereof.
  • agonistic is used herein interchangeably with the term “agonist”.
  • an isolated antibody or an antigen-binding fragment thereof, that binds to LAG-3 and inhibits antigen-induced CD4 + and/or CD8 + T cell proliferation, or antigen-induced CD4 + and/or CD8 + T cell activation.
  • the antibody, or antigen-binding fragment thereof inhibits antigen-induced CD4 + T cell proliferation, and/or antigen-induced CD8 + T cell proliferation. In some embodiments, the antibody, or antigen-binding fragment thereof, inhibits antigen-induced CD4 + T cell proliferation, and antigen-induced CD8 + T cell proliferation. In particular embodiments, the antibody, or antigen-binding fragment thereof, inhibits antigen-induced CD8 + T cell proliferation more than antigen-induced CD4 + T cell proliferation.
  • FIG. 21 illustrates the differences between depleting anti-LAG-3 antibodies, antagonist anti-LAG-3 antibodies, and antibodies of the invention (i.e. agonistic anti-LAG-3 antibodies, and antibodies that bind to LAG-3 and inhibit antigen-induced CD4 + and/or CD8 + T cell proliferation, or antigen-induced CD4 + and/or CD8 + T cell activation).
  • a depleting anti-LAG-3 antibody causes depletion of activated T cells by binding to LAG-3 expressed on the surface of the cells. Depletion can occur by antibody-dependent cell-mediated cytotoxicity (ADCC), or by complement-dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • the Fc region of the depleting antibody binds to Fc receptors (Fc ⁇ Rs) on the surface of immune effector cells, such as natural killers and macrophages, leading to lysis of the targeted cells.
  • Fc ⁇ Rs Fc receptors
  • CDC the Fc region of the depleting antibody binds to the C1q component of complement, and the targeted cell is killed by triggering the complement cascade at the cell surface.
  • depleting anti-LAG-3 antibodies inhibit T cell-mediated immune responses. It will be appreciated that the effects of depleting anti-LAG-3 antibodies are long-lasting, and irreversible because they cause the destruction of activated T cells.
  • An antagonist anti-LAG-3 antibody binds to LAG-3 on the surface of activated T cells, and prevents interaction of LAG-3 with MHC class II molecules on the surface of antigen-presenting cells (APCs). This blocks the negative regulation of signal transduction that occurs when APCs bind to LAG-3 on activated T cells. Consequently, antagonist anti-LAG-3 antibodies prevent the negative regulation of T cell proliferation, function and homeostasis normally mediated by LAG-3. Such antibodies are useful, for example, for the treatment of cancer and infectious disease.
  • Antibodies of the invention bind to LAG-3 on the surface of activated T cells, and negatively regulate signal transduction through agonism of LAG-3, causing negative regulation of T cell proliferation and/or activation.
  • antibodies of the invention inhibit T cell-mediated immune responses, in particular by inhibiting antigen-induced CD4 + and/or CD8 + T cell proliferation, and/or antigen-induced CD4 + and/or CD8 + T cell activation.
  • the effects of such antibodies are reversible, and may be less long-lasting than the effects of depleting anti-LAG-3 antibodies since they do not cause the destruction of activated T cells. It will be appreciated that the length of time for which an antibody of the invention is effective will depend on the plasma half-life of the antibody.
  • Inhibition of antigen-induced CD4 + and/or CD8 + T cell proliferation may be determined by any suitable method known to the skilled person.
  • An example of a suitable method is by measuring the proliferation of CD4 + and/or CD8 + T cells induced by antigenic peptides in the presence of the antibody or fragment, compared with the corresponding proliferation in the presence of a negative control antibody of the same isotype.
  • the CD4 + and CD8 + T cells may be present, for example, in a sample of peripheral blood mononuclear cells (PBMCs) obtained from a healthy donor.
  • Proliferation of the cells may be induced by any suitable antigenic peptides, such as a pool of peptides covering the sequence of CMV pp35.
  • Proliferation of the cells may be measured by labelling the cells, for example with a fluorescent cell staining dye, such as carboxyfluorescein succinimidyl ester (CFSE).
  • CFSE carboxyfluorescein succinimidyl ester
  • the percentage inhibition of antigen-induced CD4 + and/or CD8 + T cell proliferation may be determined as a percentage inhibition of the proliferation index (PI), calculated as the sum of: the percentage of CD4 + and/or CD8 + T cells under each division peak (assessed by FACS), multiplied by the division number, as described in more detail in Example 10 below.
  • PI percentage inhibition of the proliferation index
  • an antibody of the invention inhibits antigen-induced CD4 + T cell proliferation by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%, compared to antigen-induced CD4 + T cell proliferation in the absence of the antibody or fragment.
  • an antibody of the invention inhibits antigen-induced CD8 + T cell proliferation by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%, compared to antigen-induced CD8 + T cell proliferation in the absence of the antibody or fragment.
  • an antibody of the invention inhibits antigen-induced CD4 + T cell proliferation, and antigen-induced CD8 + T cell proliferation, each by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%, compared to antigen-induced CD4 + T cell proliferation, and antigen-induced CD8 + T cell proliferation, respectively, in the absence of the antibody or fragment.
  • an antibody of the invention inhibits antigen-induced CD4 + T cell proliferation by at least 20% compared to antigen-induced CD4 + T cell proliferation in the absence of the antibody or fragment, and inhibits antigen-induced CD8 + T cell proliferation by at least 30% compared to and antigen-induced CD8 + T cell proliferation in the absence of the antibody or fragment.
  • Inhibition of antigen-induced CD4 + and/or CD8 + T cell proliferation by an antibody of the invention, or fragment thereof, may be compared to antigen-induced CD4 + and/or CD8 + T cell proliferation in the presence of a negative control antibody of the same isotype, or fragment thereof.
  • an antibody of the invention inhibits antigen-induced CD8 + T cell proliferation by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%, more than the antibody or fragment inhibits antigen-induced CD4 + T cell proliferation.
  • the inhibition of antigen-induced CD8 + T cell proliferation is LAG-3 dependent and IL-2 independent.
  • an antibody of the invention, or antigen-binding fragment thereof inhibits antigen-induced CD4 + and/or CD8 + T cell activation. In certain embodiments, an antibody of the invention, or antigen-binding fragment thereof, inhibits antigen-induced CD4 + and/or CD8 + T cell proliferation and antigen-induced CD4 + and/or CD8 + T cell activation.
  • an antibody of the invention, or antigen-binding fragment thereof may bind to LAG-3 and inhibit antigen-induced CD4 + and/or CD8 + T cell activation.
  • an antibody of the invention, or antigen-binding fragment thereof binds to LAG-3 and inhibits antigen-induced CD4 + T cell activation and antigen-induced CD8 + T cell activation.
  • Inhibition of activation of CD4 + and/or CD8 + T cells may be determined by any suitable method known to the skilled person.
  • An example of a suitable method is by measuring the effect of the antibody, or fragment, on CD4 + and/or CD8 + T cell activation marker expression, or T cell activation marker secretion.
  • CD8 + T cell activation may be measured by measuring the expression of CD25, as an activation marker, on CD8 + T cells induced by antigenic peptides in the presence of the antibody or fragment, compared with the corresponding CD25 expression in the presence of a negative control antibody of the same isotype.
  • T cell activation may be measured by measuring the secretion of IFN- ⁇ in cell supernatant of T cells induced by antigenic peptides in the presence of the antibody or fragment, compared with the corresponding secretion in the presence of a negative control antibody of the same isotype.
  • the T cells may be present, for example, in a sample of PBMCs obtained from a healthy donor. Activation of the cells may be induced by any suitable antigenic peptides, such as a pool of peptides covering the sequence of CMV pp35.
  • An example of a method for determining inhibition of antigen-induced T cell activation by measuring T cell activation marker secretion is described in more detail in Example 15 below.
  • An example of a method for determining inhibition of antigen-induced CD8 + T cell activation by measuring CD8 + T cell activation marker expression is described in more detail in Example 16 below.
  • an antibody of the invention inhibits antigen-induced CD4 + and/or CD8 + T cell activation by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%, compared to antigen-induced CD4 + and/or CD8 + T cell activation in the absence of the antibody or fragment.
  • Inhibition of antigen-induced CD4 + and/or CD8 + T cell activation by an antibody of the invention, or fragment thereof, may be compared to antigen-induced CD4 + and/or CD8 + T cell activation in the presence of a negative control antibody of the same isotype, or fragment thereof.
  • an antibody of the invention inhibits binding of IMP321 to MHC class II-positive cells.
  • IMP321 (also referred to as “LAG-3Ig” below) is a recombinant soluble human LAG-3Ig fusion protein.
  • the fusion protein is obtained as a 200-kDa dimer produced in Chinese hamster ovary (CHO) cells transfected with a plasmid encoding for the extracellular domain of human LAG-3 fused to the human IgG1 Fc.
  • the sequence of IMP321 is provided in SEQ ID NO: 17 of US Patent Application No. 2011/0008331.
  • Binding of IMP321 to MHC class II-positive cells may be determined by measuring binding of an IMP321-label conjugate (for example an IMP321-Alex 488 conjugate) to Raji cells (these are MHC class II-positive B cells), for example as described in Example 8 below.
  • an IMP321-label conjugate for example an IMP321-Alex 488 conjugate
  • Raji cells these are MHC class II-positive B cells
  • an antibody of the invention inhibits binding of IMP321 to MHC class II-positive cells by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% compared to the binding of IMP321 to MHC class II-positive cells in the absence of the antibody or fragment.
  • an antibody of the invention inhibits binding of IMP321 to MHC class II-positive cells by at least 30%, compared to the binding of IMP321 to MHC class II-positive cells in the absence of the antibody or fragment, wherein the ratio of the concentration of the antibody, or fragment, to IMP321 is 0.1:1.
  • an antibody of the invention inhibits binding of IMP321 to MHC class II-positive cells by at least 80%, compared to the binding of IMP321 to MHC class II-positive cells in the absence of the antibody or fragment, wherein the ratio of the concentration of the antibody, or fragment, to IMP321 is 0.3:1 or 1:1.
  • an antibody of the invention inhibits IMP321-induced monocyte activation.
  • IMP321 is able to activate cells of the human monocytic cell line THP-1. Activation of THP-1 cells can be determined by the level of secretion of chemokine ligand 4 (CCL4, also known as Macrophage inflammatory protein-1 ⁇ , MIP-1 ⁇ ) by the THP-1 cells. Pre-incubation of an antibody, or fragment, of the invention with IMP321 prior to incubation of the mixture with THP-1 cells can be used to determine whether the antibody, or fragment, inhibits IMP321-induced monocyte activation. A method for determining inhibition of IMP321-induced monocyte activation is described in more detail in Example 9 below.
  • an antibody of the invention inhibits IMP321-induced monocyte activation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% compared to the amount of IMP321-induced monocyte activation in the absence of the antibody or fragment.
  • an antibody of the invention inhibits IMP321-induced monocyte activation by at least 70% compared to the amount of IMP321-induced monocyte activation in the absence of the antibody or fragment, wherein the ratio of the concentration of the antibody, or fragment, to IMP321 is 1:1.
  • Huard et al Proc. Natl. Acad. Sci. USA, 11: 5744-5749, 1997) describes characterization of the MHC class II binding site on LAG-3 protein. Many of the residues essential for binding MHC class II proteins are clustered at the base of a large 30 amino acid extra-loop structure in the LAG-3 D1 domain.
  • the amino acid sequence of the extra-loop structure of the D1 domain of human LAG-3 protein is GPPAAAPGHPLAPGPHPAAPSSWGPRPRRY (SEQ ID NO: 40), the underlined sequence in FIG. 1 .
  • An antibody of the invention may bind to an epitope of human LAG-3 that overlaps with the MHC class II binding site on LAG-3.
  • An antibody of the invention may bind to an epitope that overlaps with the 30 amino acid extra-loop of the first N-terminal D1 domain of human LAG-3.
  • an antibody of the invention does not bind to the 30 amino acid extra-loop sequence (SEQ ID NO: 40) of the first N-terminal D1 domain of human LAG-3 protein.
  • An antibody of the invention may inhibit binding of LAG-3 to MHC class II molecules in vivo.
  • an antibody of the invention may antagonise the MHC class II-activating signal into antigen-presenting cells (APCs).
  • APCs antigen-presenting cells
  • an antibody of the invention may inhibit LAG-3-induced APC activation, for example dendritic cell activation, for example LAG-3-induced monocyte or macrophage activation.
  • an antibody of the invention inhibits binding of LAG-3 to MHC class II-positive cells by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% compared to the binding of LAG-3 to MHC class II-positive cells in the absence of the antibody or fragment.
  • an antibody of the invention inhibits LAG-3-induced APC activation by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% compared to the amount of LAG-3-induced APC activation in the absence of the antibody or fragment.
  • a monoclonal antibody of the invention, or antigen-binding fragment thereof may comprise one, two, or three complementarity determining regions (CDRs) of an antibody heavy chain variable (VH) region comprising amino acid sequence SEQ ID NO:7, and/or one, two, or three CDRs of an antibody light chain variable (VL) region comprising amino acid sequence SEQ ID NO:8.
  • CDRs complementarity determining regions
  • an anti-LAG-3 antibody, or antigen-binding fragment thereof which comprises one, two, or three complementarity determining regions (CDRs) of an antibody heavy chain variable (VH) region comprising amino acid sequence SEQ ID NO:7, and/or one, two, or three CDRs of an antibody light chain variable (VL) region comprising amino acid sequence SEQ ID NO:8.
  • CDRs complementarity determining regions
  • the CDRs of the antibody VH region may be CDRs of amino acid sequence SEQ ID NO:1, 2, and 3, and the CDRs of the antibody VL region may be CDRs of amino acid sequence SEQ ID NO:4, 5, and 6.
  • An antibody of the invention may comprise an antibody VH region with CDRs comprising amino acid sequences of SEQ ID NOs:1, 2, and 3, and/or an antibody VL region with CDRs comprising amino acid sequences of SEQ ID NOs:4, 5, and 6.
  • the CDRs of amino acid sequence SEQ ID NOs:1, 2, and 3 may be present in any order in the VH region, and the CDRs of amino acid sequence SEQ ID NOs:4, 5, and 6 may be present in any order in the VL region.
  • the antibody, or fragment thereof comprises CDR-H1 having amino acid sequence SEQ ID NO:1, CDR-H2 having amino acid sequence SEQ ID NO:2, and CDR-H3 having amino acid sequence SEQ ID NO:3, and/or CDR-L1 having amino acid sequence SEQ ID NO:4, CDR-L2 having amino acid sequence SEQ ID NO:5, and CDR-L3 having amino acid sequence SEQ ID NO:6.
  • the CDRs of the antibody VH region may be CDRs of amino acid sequence SEQ ID NO:21, 22, and 23, and the CDRs of the antibody VL region may be CDRs of amino acid sequence SEQ ID NO:24, 25, and 26.
  • An antibody of the invention, or antigen-binding fragment thereof may comprise an antibody VH region with CDRs comprising amino acid sequences of SEQ ID NOs:21, 22, and 23, and/or an antibody VL region with CDRs comprising amino acid sequences of SEQ ID NOs:24, 25, and 26.
  • the CDRs of amino acid sequence SEQ ID NOs:21, 22, and 23 may be present in any order in the VH region, and the CDRs of amino acid sequence SEQ ID NOs:24, 25, and 26 may be present in any order in the VL region.
  • the antibody, or fragment thereof comprises CDR-H1 having amino acid sequence SEQ ID NO:21, CDR-H2 having amino acid sequence SEQ ID NO:22, and CDR-H3 having amino acid sequence SEQ ID NO:23, and/or CDR-L1 having amino acid sequence SEQ ID NO:24, CDR-L2 having amino acid sequence SEQ ID NO:25, and CDR-L3 having amino acid sequence SEQ ID NO:26.
  • the CDRs of the antibody VH region are selected from CDRs of amino acid sequence SEQ ID NO:1, 2, 3, 21, 22, and 23, and the CDRs of the antibody VL region are selected from CDRs of amino acid sequence SEQ ID NO:4, 5, 6, 24, 25, and 26.
  • an antibody of the invention comprises an antibody VH region comprising a VH CDR1, a VH CDR2, and a VH CDR3, wherein the VH CDR1 has an amino acid sequence selected from SEQ ID NO:1 and 21, and/or the VH CDR2 has an amino acid sequence selected from SEQ ID NO:2 and 22, and/or the VH CDR3 has an amino acid sequence selected from SEQ ID NO:3 and 23.
  • an antibody of the invention comprises an antibody VL region comprising a VL CDR1, a VL CDR2, and a VL CDR3, wherein the VL CDR1 has an amino acid sequence selected from SEQ ID NO:4 and 24, and/or the VL CDR2 has an amino acid sequence selected from SEQ ID NO:5 and 25, and/or the VL CDR3 has an amino acid sequence selected from SEQ ID NO:6 and 26.
  • an antibody of the invention comprises: an antibody VH region comprising: a VH CDR1 with an amino acid sequence selected from SEQ ID NO:1 and 21; a VH CDR2 with an amino acid sequence selected from SEQ ID NO:2 and 22; and a VH CDR3 with an amino acid sequence selected from SEQ ID NO:3 and 23; and an antibody VL region comprising: a VL CDR1 with an amino acid sequence selected from SEQ ID NO:4 and 24; a VL CDR2 with an amino acid sequence selected from SEQ ID NO:5 and 25; and a VL CDR3 with an amino acid sequence selected from SEQ ID NO:6 and 26.
  • An antibody of the invention, or antigen-binding fragment thereof may comprise an antibody VH region comprising amino acid sequence that is at least 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO:7, and/or an antibody VL region comprising amino acid sequence that is at least 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO:8.
  • an antibody of the invention comprises an antibody VH region comprising amino acid sequence SEQ ID NO:7, and/or an antibody VL region comprising amino acid sequence SEQ ID NO:8.
  • An antibody of the invention may comprise an antibody VH region and/or an antibody VL region comprising amino acid sequence that is at least 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical, or that is identical, to the amino acid sequence of the VH and/or VL regions of mouse monoclonal anti-LAG-3 antibody 13E2, described herein in Examples 1, 2 and 3 (13E2 VH amino acid sequence: SEQ ID NO:7; 13E2 VL amino acid sequence: SEQ ID NO:8).
  • a monoclonal antibody of the invention, or antigen-binding fragment thereof may comprise one, two, or three complementarity determining regions (CDRs) of an antibody heavy chain variable (VH) region comprising amino acid sequence SEQ ID NO:17, and/or one, two, or three CDRs of an antibody light chain variable (VL) region comprising amino acid sequence SEQ ID NO:18.
  • CDRs complementarity determining regions
  • an anti-LAG-3 antibody, or antigen-binding fragment thereof which comprises one, two, or three complementarity determining regions (CDRs) of an antibody heavy chain variable (VH) region comprising amino acid sequence SEQ ID NO:17, and/or one, two, or three CDRs of an antibody light chain variable (VL) region comprising amino acid sequence SEQ ID NO:18.
  • CDRs complementarity determining regions
  • the CDRs of the antibody VH region may be CDRs of amino acid sequence SEQ ID NO:11, 12, and 13 and the CDRs of the antibody VL region may be CDRs of amino acid sequence SEQ ID NO:14, 15, and 16.
  • An antibody of the invention, or antigen-binding fragment thereof may comprise an antibody VH region with CDRs comprising amino acid sequences of SEQ ID NOs:11, 12, and 13, and/or an antibody VL region with CDRs comprising amino acid sequences of SEQ ID NOs:14, 15, and 16.
  • the CDRs of amino acid sequence SEQ ID NOs:11, 12, and 13 may be present in any order in the VH region, and the CDRs of amino acid sequence SEQ ID NOs:14, 15, and 16 may be present in any order in the VL region.
  • the antibody, or fragment thereof comprises CDR-H1 having amino acid sequence SEQ ID NO:11, CDR-H2 having amino acid sequence SEQ ID NO:12, and CDR-H3 having amino acid sequence SEQ ID NO:13, and/or CDR-L1 having amino acid sequence SEQ ID NO:14, CDR-L2 having amino acid sequence SEQ ID NO:15, and CDR-L3 having amino acid sequence SEQ ID NO:16.
  • the CDRs of the antibody VH region may be CDRs of amino acid sequence SEQ ID NO:31, 32, and 33
  • the CDRs of the antibody VL region may be CDRs of amino acid sequence SEQ ID NO:34, 35, and 36.
  • An antibody of the invention, or antigen-binding fragment thereof may comprise an antibody VH region with CDRs comprising amino acid sequences of SEQ ID NOs:31, 32, and 33, and/or an antibody VL region with CDRs comprising amino acid sequences of SEQ ID NOs:34, 35, and 36.
  • the CDRs of amino acid sequence SEQ ID NOs:31, 32, and 33 may be present in any order in the VH region, and the CDRs of amino acid sequence SEQ ID NOs:34, 35, and 36 may be present in any order in the VL region.
  • the antibody, or fragment thereof comprises CDR-H1 having amino acid sequence SEQ ID NO:31, CDR-H2 having amino acid sequence SEQ ID NO:32, and CDR-H3 having amino acid sequence SEQ ID NO:33, and/or CDR-L1 having amino acid sequence SEQ ID NO:34, CDR-L2 having amino acid sequence SEQ ID NO:35, and CDR-L3 having amino acid sequence SEQ ID NO:36.
  • the CDRs of the antibody VH region are selected from CDRs of amino acid sequence SEQ ID NO:11, 12, 13, 31, 32, and 33, and the CDRs of the antibody VL region are selected from CDRs of amino acid sequence SEQ ID NO:14, 15, 16, 34, 35, and 36.
  • an antibody of the invention comprises an antibody VH region comprising a VH CDR1, a VH CDR2, and a VH CDR3, wherein the VH CDR1 has an amino acid sequence selected from SEQ ID NO:11 and 31, and/or the VH CDR2 has an amino acid sequence selected from SEQ ID NO:12 and 32, and/or the VH CDR3 has an amino acid sequence selected from SEQ ID NO:13 and 33.
  • an antibody of the invention comprises an antibody VL region comprising a VL CDR1, a VL CDR2, and a VL CDR3, wherein the VL CDR1 has an amino acid sequence selected from SEQ ID NO:14 and 34, and/or the VL CDR2 has an amino acid sequence selected from SEQ ID NO:15 and 35, and/or the VL CDR3 has an amino acid sequence selected from SEQ ID NO:16 and 36.
  • an antibody of the invention comprises: an antibody VH region comprising: a VH CDR1 with an amino acid sequence selected from SEQ ID NO:11 and 31, a VH CDR2 with an amino acid sequence selected from SEQ ID NO:12 and 32, and a VH CDR3 with an amino acid sequence selected from SEQ ID NO:13 and 33; and an antibody VL region comprising: a VL CDR1 with an amino acid sequence selected from SEQ ID NO:14 and 34, a VL CDR2 with an amino acid sequence selected from SEQ ID NO:15 and 35, and a VL CDR3 with an amino acid sequence selected from SEQ ID NO:16 and 36.
  • An antibody of the invention, or antigen-binding fragment thereof may comprise an antibody VH region comprising amino acid sequence that is at least 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO:17, and/or an antibody VL region comprising amino acid sequence that is at least 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO:18.
  • an antibody of the invention comprises an antibody VH region comprising amino acid sequence SEQ ID NO:17, and/or an antibody VL region comprising amino acid sequence SEQ ID NO:18.
  • An antibody of the invention may comprise an antibody VH region and an antibody VL region comprising amino acid sequence that is at least 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical, or that is identical, to the amino acid sequence of the VH and VL regions of mouse monoclonal anti-LAG-3 antibody 34F4, described herein in Examples 4, 5 and 6 (34F4 VH amino acid sequence: SEQ ID NO:17; 34F4 VL amino acid sequence: SEQ ID NO:18).
  • antibodies of the invention comprise a heavy chain variable region comprising CDR1, CDR2 and CDR3 sequences, and a light chain variable region comprising CDR1, CDR2, and CDR3 sequences, which differ from those of antibody 13E2 or 34F4 by one or more conservative modifications, for example up to five conservative modifications. It is understood in the art that certain conservative sequence modifications can be made which do not remove antigen binding. See, e.g., Brummell et al. (1993) Biochem 32:1180-8; de Wildt et al. (1997) Prot. Eng. 10:835-41; Komissarov et al. (1997) J. Biol. Chem. 272:26864-26870; Hall et al.
  • conservative sequence modifications refers to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody of the invention by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • amino acid residues within the CDR regions of an antibody of the invention can be replaced with other amino acid residues from the same side chain family and the altered antibody can be tested for retained function (i.e., the functions set forth above) using the functional assays described herein.
  • Antibodies of the invention can be prepared using an antibody having one or more of the VH and/or VL sequences of 13E2 or 34F4 as starting material to engineer a modified antibody.
  • An antibody can be engineered by modifying one or more residues within one or both variable regions (i.e., VH and/or VL), for example within one or more CDR regions and/or within one or more framework regions. Additionally or alternatively, an antibody can be engineered by modifying residues within the constant region(s), for example to alter the effector function(s) of the antibody.
  • CDR grafting can be used to engineer variable regions of antibodies.
  • Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to express recombinant antibodies that mimic the properties of specific naturally occurring antibodies by constructing expression vectors that include CDR sequences from the specific naturally occurring antibody grafted onto framework sequences from a different antibody with different properties (see, e.g., Riechmann et al. (1998) Nature 332:323-327; Jones et al.
  • an antibody of the invention for example a monoclonal antibody, or antigen-binding fragment thereof, comprising CDR1, CDR2, and CDR3 of an antibody heavy chain variable region comprising amino acid sequence SEQ ID NO:7, and/or comprising CDR1, CDR2, and CDR3 of an antibody light chain variable region comprising amino acid sequence SEQ ID NO:8 (i.e., the CDRs of 13E2). While such antibodies contain the VH and VL CDR sequences of monoclonal antibody 13E2, they can contain differing framework sequences.
  • an antibody of the invention for example a monoclonal antibody, or antigen-binding fragment thereof, comprising CDR1, CDR2, and CDR3 of an antibody heavy chain variable region comprising amino acid sequence SEQ ID NO:17, and/or comprising CDR1, CDR2, and CDR3 of an antibody light chain variable region comprising amino acid sequence SEQ ID NO:18 (i.e., the CDRs of 34F4). While such antibodies contain the VH and VL CDR sequences of monoclonal antibody 34F4, they can contain differing framework sequences.
  • Such framework sequences can be obtained from public DNA databases or published references that include germline antibody gene sequences.
  • germline DNA sequences for human heavy and light chain variable region genes can be found in the “VBase” human germline sequence database (available on the Internet at www.mrc-cpe.cam.ac.uk/vbase), as well as in Kabat et al. (1991), cited supra; Tomlinson et al. (1992) “The Repertoire of Human Germline V H Sequences Reveals about Fifty Groups of V H Segments with Different Hypervariable Loops” J. Mol. Biol. 227:776-798; and Cox et al.
  • the germline DNA sequences for human heavy and light chain variable region genes can be found in the Genbank database.
  • the following heavy chain germline sequences found in the HCo7 HuMAb mouse are available in the accompanying Genbank Accession Nos.: 1-69 (NG_0010109, NT_024637 & BC070333), 3-33 (NG_0010109 & NT_024637) and 3-7 (NG_0010109 & NT_024637).
  • Antibody protein sequences are compared against a compiled protein sequence database using one of the sequence similarity searching methods called the Gapped BLAST (Altschul et al. (1997), supra), which is well known to those skilled in the art.
  • Preferred framework sequences for use in antibodies of the invention are those that are structurally similar to the framework sequences of the 13E2 or 34F4 antibodies.
  • Sequences showing significant alignment with nucleic acid sequence encoding the VH domain of monoclonal antibody 13E2 include the following germline genes: IGHV8-8*01, IGHV8-11*01, IGHV8-12*01, IGHD2-12*01, IGHD1-1*01, IGHJ1*01, IGHJ1*02, IGHJ1*03.
  • Sequences showing significant alignment with nucleic acid sequence encoding the VL domain of monoclonal antibody 13E2 include the following germline genes: IGKV6-17*01, IGKV6-25*01, IGKV6-23*01, IGKJ2*01, IGKJ2*03, IGKJ2*02.
  • Sequences showing significant alignment with nucleic acid sequence encoding the VH domain of monoclonal antibody 34F4 include the following germline genes: IGHV8-8*01, IGHV8-12*01, IGHV8-11*01, IGHD1-1*01, IGHD1-2*01, IGHD2-3*01, IGHJ2*01, IGHJ2*02, IGHJ2*03.
  • Sequences showing significant alignment with nucleic acid sequence encoding the VL domain of monoclonal antibody 34F4 include the following germline genes: IGKV6-17*01, IGKV6-25*01, IGKV6-23*01, IGKJ1*01, IGKJ1*02, IGKJ2*01.
  • Preferred heavy chain framework sequences for use in antibodies of the invention are those that are structurally similar to the framework sequences encoded by germline V gene IGHV8-8*01, IGHV8-11*01, or IGHV8-12*01, especially IGHV8-8*01.
  • Preferred light chain framework sequences for use in antibodies of the invention are those that are structurally similar to the framework sequences encoded by germline V gene IGKV6-17*01, IGKV6-25*01, or IGKV6-23*01, especially IGKV6-17*01.
  • the VH CDR1, CDR2, and CDR3 sequences, and the VL CDR1, CDR2, and CDR3 sequences can be grafted onto framework regions that have the identical sequence as that found in the germline immunoglobulin gene from which the framework sequence derive, or the CDR sequences can be grafted onto framework regions that contain one or more mutations as compared to the germline sequences. For example, it has been found that in certain instances it is beneficial to mutate residues within the framework regions to maintain or enhance the antigen binding ability of the antibody (see e.g., U.S. Pat. Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370).
  • variable region modification is to mutate amino acid residues within the VH and/or VL CDR1, CDR2 and/or CDR3 regions to thereby improve one or more binding properties (e.g., affinity) of the antibody of interest.
  • Site-directed mutagenesis or PCR-mediated mutagenesis can be performed to introduce the mutation(s) and the effect on antibody binding, or other functional property of interest, can be evaluated in in vitro or in vivo assays as described herein and provided in the Examples.
  • Preferably conservative modifications are introduced.
  • the mutations can be amino acid substitutions, additions or deletions, but are preferably substitutions.
  • typically no more than one, two, three, four or five residues within a CDR region are altered. In some embodiments, no more than one, two, three, four or five residues are altered in total for all six CDR regions.
  • an antibody of the invention or antigen-binding fragment thereof, comprises:
  • an antibody VH region comprising: a VH CDR1 with an amino acid sequence selected from SEQ ID NO:1 and 21; a VH CDR2 with an amino acid sequence selected from SEQ ID NO:2 and 22; and a VH CDR3 with an amino acid sequence selected from SEQ ID NO:3 and 23; and an antibody VL region comprising: a VL CDR1 with an amino acid sequence selected from SEQ ID NO:4 and 24; a VL CDR2 with an amino acid sequence selected from SEQ ID NO:5 and 25; and a VL CDR3 with an amino acid sequence selected from SEQ ID NO:6 and 26; or
  • an antibody of the invention or antigen-binding fragment thereof, comprises:
  • an antibody VH region comprising: a VH CDR1 with an amino acid sequence selected from SEQ ID NO:11 and 31, a VH CDR2 with an amino acid sequence selected from SEQ ID NO:12 and 32, and a VH CDR3 with an amino acid sequence selected from SEQ ID NO:13 and 33; and an antibody VL region comprising: a VL CDR1 with an amino acid sequence selected from SEQ ID NO:14 and 34, a VL CDR2 with an amino acid sequence selected from SEQ ID NO:15 and 35, and a VL CDR3 with an amino acid sequence selected from SEQ ID NO:16 and 36; or
  • the invention provides an anti-LAG-3 monoclonal antibody, or antigen-binding fragment thereof, comprising a heavy chain variable region comprising: (a) a VH CDR1 region comprising SEQ ID NO: 1, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 1; (b) a VH CDR2 region comprising SEQ ID NO: 2, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 2; and (c) a VH CDR3 region comprising SEQ ID NO: 3, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 3; and/or a light chain variable region comprising: (a) a VL CDR1 region comprising SEQ ID NO: 4, or an amino acid sequence having one, two, three, four or five amino acid substitutions
  • the invention provides an anti-LAG-3 monoclonal antibody, or antigen-binding fragment thereof, comprising a heavy chain variable region comprising: (a) a VH CDR1 region comprising SEQ ID NO: 11, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 11; (b) a VH CDR2 region comprising SEQ ID NO: 12, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 12; and (c) a VH CDR3 region comprising SEQ ID NO: 13, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 13; and/or a light chain variable region comprising: (a) a VL CDR1 region comprising SEQ ID NO: 14, or an amino acid sequence having one, two, three, four or five amino acid
  • the invention provides an anti-LAG-3 monoclonal antibody, or antigen-binding fragment thereof, comprising a heavy chain variable region comprising: (a) a VH CDR1 region comprising SEQ ID NO: 21, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 21; (b) a VH CDR2 region comprising SEQ ID NO: 22, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 22; and (c) a VH CDR3 region comprising SEQ ID NO: 23, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 23; and/or a light chain variable region comprising: (a) a VL CDR1 region comprising SEQ ID NO: 24, or an amino acid sequence having one, two, three, four or five amino acid substitutions
  • the invention provides an anti-LAG-3 monoclonal antibody, or antigen-binding fragment thereof, comprising a heavy chain variable region comprising: (a) a VH CDR1 region comprising SEQ ID NO: 31, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 31; (b) a VH CDR2 region comprising SEQ ID NO: 32, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 32; and (c) a VH CDR3 region comprising SEQ ID NO: 33, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NO: 33; and/or a light chain variable region comprising: (a) a VL CDR1 region comprising SEQ ID NO: 34, or an amino acid sequence having one, two, three, four or
  • Engineered antibodies of the invention include those in which modifications have been made to framework residues within VH and/or VL, e.g. to improve the properties of the antibody. Typically such framework modifications are made to decrease the immunogenicity of the antibody. For example, one approach is to “backmutate” one or more framework residues to the corresponding germline sequence. More specifically, an antibody that has undergone somatic mutation can contain framework residues that differ from the germline sequence from which the antibody is derived. Such residues can be identified by comparing the antibody framework sequences to the germline sequences from which the antibody is derived.
  • Another type of framework modification involves mutating one or more residues within the framework region, or even within one or more CDR regions, to remove T cell epitopes to thereby reduce the potential immunogenicity of the antibody. This approach is also referred to as “deimmunization” and is described in further detail in U.S. Patent Publication No. 20030153043.
  • antibodies of the invention can be engineered to include modifications within the Fc region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or antigen-dependent cellular cytotoxicity.
  • an antibody of the invention can be chemically modified (e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation, again to alter one or more functional properties of the antibody.
  • the hinge region of CF11 is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. This approach is described further in U.S. Pat. No. 5,677,425. The number of cysteine residues in the hinge region of CF11 is altered to, for example, facilitate assembly of the light and heavy chains or to increase or decrease the stability of the antibody.
  • the Fc hinge region of an antibody is mutated to decrease the biological half-life of the antibody. More specifically, one or more amino acid mutations are introduced into the CF12-CF13 domain interface region of the Fc-hinge fragment such that the antibody has impaired Staphylococcyl protein A (SpA) binding relative to native Fc-hinge domain SpA binding.
  • SpA Staphylococcyl protein A
  • the antibody is modified to increase its biological half-life.
  • the IgG class is the most stable and has a serum half-life of 20 days, whereas IgM and IgA persist for only 5-8 days (Brekke & Sandlie, 2003, Nature Reviews Drug Discovery 2, 52-62).
  • Various approaches are possible. For example, one or more of the following mutations can be introduced: T252L, T254S, T256F, as described in U.S. Pat. No. 6,277,375.
  • the antibody can be altered within the CF11 or CL region to contain a salvage receptor binding epitope taken from two loops of a CF12 domain of an Fc region of an IgG, as described in U.S. Pat. Nos. 5,869,046 and 6,121,022.
  • An antibody of the invention should lack antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) so that the antibody or fragment can be used to negatively regulate T cell proliferation and/or function without depleting T cells as a result of ADCC or CDC.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • ADCC the Fc region of an antibody binds to Fc receptors (Fc ⁇ Rs) on the surface of immune effector cells, such as natural killers and macrophages, leading to lysis of the targeted cells.
  • Fc ⁇ Rs Fc receptors
  • CDC the Fc region binds to the C1q component of complement, and the targeted cell is killed by triggering the complement cascade at the cell surface.
  • the ADCC and CDC activity of an antibody depends on its isotype. Both IgM and IgG can mediate complement fixation, whereas only IgG can promote antibody-dependent cellular cytotoxicity (ADCC). IgG isoforms exhibit different levels of CDC and ADCC:
  • an antibody of the invention comprises a mouse IgG1, or human IgG4 Fc portion to ensure that the antibody or fragment lacks ADCC and CDC activity.
  • ADCC and CDC activity of an antibody of the invention, or antigen-binding fragment thereof may be determined by any suitable method known to the skilled person. Examples of suitable assays for CDC and ADCC activity are described in WO 2008/132601, and below.
  • An anti-LAG-3 antibody exhibiting CDC activity will consistently kill LAG-3+ cells in the presence of complement, compared with its isotypic control.
  • the target cells used to assess an anti-LAG-3 antibody may be a LAG-3-transfected cell line, or primary T cells activated to induce expression of LAG-3.
  • the target cells are LAG-3+ CHO cells compared to wt CHO cells. Both types of cells (i.e. cells expressing LAG-3, and equivalent cells not expressing LAG-3) are incubated for 1 hour at 37° C. with either the anti-LAG-3 test antibody, or its isotype-matched negative control antibody, and rabbit serum containing active complement.
  • 7-Amino-Actinomycin D 7-Amino-Actinomycin D
  • 7-AAD 7-Amino-Actinomycin D
  • the percentage of 7-AAD-positive CHO cells is determined by flow cytometry analysis.
  • An antibody exhibiting CDC activity will only kill LAG-3+ cells (for example, LAG-3+ CHO cells) in the presence of complement.
  • the anti-LAG-3 Ab may be titered down to determine the efficacy of the antibody to activate CDC at low concentration of antibody.
  • a CDC assay may also be performed on PBMCs stimulated with the superantigen SEB.
  • the cytotoxicity of test antibody is analysed on both activated (namely CD25+/LAG-3+ cells) and non-activated (namely CD25 ⁇ /LAG-3 ⁇ cells) CD4+ helper T and CD8+ cytotoxic T cells. Only activated CD4 + and CD8 + T cells are specifically killed by an antibody exhibiting CDC.
  • test anti-LAG-3 antibody if effector cells (PBMCs) are able to kill a significant percentage of LAG-3-expressing cells (for example, LAG-3+ CHO cells), compared with an isotype-matched negative control antibody, the test antibody exhibits ADCC. This effect should increase with the number of effector cells.
  • the test antibody may be titered down to determine the efficacy of the antibody to induce ADCC at low concentration of antibody.
  • test anti-LAG-3 antibody can be considered not to exhibit CDC or ADCC when it kills less than twice as many LAG-3+ cells as an isotype-matched negative control antibody in any of the assays described above.
  • ADCC bioassays which use donor peripheral blood mononuclear cells (PBMCs) or the natural killer (NK) cell subpopulation as effector cells.
  • PBMCs peripheral blood mononuclear cells
  • NK natural killer
  • An ADCC Reporter Bioassay available from Promega uses an alternative readout at an earlier point in ADCC pathway activation: the activation of gene transcription through the NFAT (nuclear factor of activated T-cells) pathway in the effector cell.
  • NFAT nuclear factor of activated T-cells
  • the ADCC Reporter Bioassay uses engineered Jurkat cells stably expressing the Fc ⁇ RIIIa receptor, V158 (high affinity) variant, and an NFAT response element driving expression of firefly luciferase as effector cells.
  • Antibody biological activity in ADCC is quantified through the luciferase produced as a result of NFAT pathway activation; luciferase activity in the effector cell is quantified with luminescence readout.
  • signal is high, and assay background is low.
  • a good assay response is only obtained when target cells with the correct surface antigen, the correct specific antibody, and effector cells expressing Fc ⁇ RIIIa are present. If any one of these is missing, there is no response.
  • the tested antibody When evaluating the ADCC activity of an antibody using an ADCC Reporter Bioassay, the tested antibody can be considered not to exhibit ADCC when the measured increase in bioluminescence is less than twice that of its isotype-matched negative control antibody.
  • the Fc region comprises a mutant human IgG4 Fc sequence with an S228P mutation to abolish Fab arm exchange (as shown in FIG. 20 (A) for chimeric antibody Chim13E2IgG4 comprising heavy chain sequence 13E2IgG4mut).
  • the Fc region comprises a wild-type human Ig kappa (IgK) chain C portion (13E2IgK) (as shown in FIG. 20 (B) for chimeric antibody Chim13E2IgG4 comprising light chain sequence 13E2IgK).
  • the numbering of residues in the Fc region used for the human IgG4 Fc mutant described above is the standard numbering of the Eu index as in Kabat (Kabat, E. A. et al., Sequences of proteins of immunological interest. 5th Edition—US Department of Health and Human Services, NIH publication n° 91-3242, pp 662, 680, 689 (1991)).
  • the glycosylation of an antibody is modified.
  • an aglycosylated antibody can be made (i.e., the antibody lacks glycosylation).
  • Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen.
  • carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence.
  • one or more amino acid substitutions can be made that result in elimination of one or more variable region framework glycosylation sites to thereby eliminate glycosylation at that site.
  • Such aglycosylation may increase the affinity of the antibody for antigen. See, e.g., U.S. Pat. Nos. 5,714,350 and 6,350,861.
  • An antibody can be pegylated to, for example, increase the biological (eg., serum) half-life of the antibody.
  • the antibody, or fragment thereof typically is reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the antibody or antibody fragment.
  • PEG polyethylene glycol
  • the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer).
  • polyethylene glycol is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (CI-C10) alkoxy- or aryloxy-polyethylene glycol or polyethylene glycol-maleimide.
  • the antibody to be pegylated is an aglycosylated antibody. Methods for pegylating proteins are known in the art and can be applied to the antibodies of the invention. See, eg., EP 0 154 316 and EP 0 401 384.
  • An antibody of the invention may be a monoclonal antibody, or an antigen-binding fragment thereof.
  • antibody and “immunoglobulin” include antibodies or immunoglobulins of any isotype, fragments of antibodies that retain specific binding to antigen, including, but not limited to, Fab, Fv, scFv, and Fd fragments, chimeric antibodies, humanized antibodies, single-chain antibodies (scAb), single domain antibodies (dAb), single domain heavy chain antibodies, a single domain light chain antibodies, bi-specific antibodies, multi-specific antibodies, and fusion proteins comprising an antigen-binding (also referred to herein as antigen binding) portion of an antibody and a non-antibody protein.
  • Fab single-chain antibodies
  • dAb single domain antibodies
  • dAb single domain heavy chain antibodies
  • bi-specific antibodies bi-specific antibodies
  • multi-specific antibodies and fusion proteins comprising an antigen-binding (also referred to herein as antigen binding) portion of an antibody and a non-antibody protein.
  • Fab′, Fv, F(ab′) 2 and or other antibody fragments that retain specific binding to antigen, and monoclonal antibodies.
  • An antibody can be monovalent or bivalent.
  • An antibody can be an Ig monomer, which is a “Y-shaped” molecule that consists of four polypeptide chains: two heavy chains and two light chains connected by disulfide bonds.
  • the antibodies can be detectably labeled, e.g., with a radioisotope, an enzyme that generates a detectable product, a fluorescent protein, and the like.
  • the antibodies can be further conjugated to other moieties, such as members of specific binding pairs, e.g., biotin (member of biotin-avidin specific binding pair), and the like.
  • the antibodies can also be bound to a solid support, including, but not limited to, polystyrene plates or beads, and the like.
  • Antibody fragments comprise a portion of an intact antibody, for example, the antigen binding or variable region of the intact antibody.
  • antibody fragments include Fab, Fab′, F(ab′) 2 , and Fv fragments; diabodies; linear antibodies (Zapata et al., Protein Eng. 8(10): 1057-1062 (1995)); domain antibodies (dAb; Holt et al. (2003) Trends Biotechnol. 21:484); single-chain antibody molecules; and multi-specific antibodies formed from antibody fragments.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called “Fab” fragments, each with a single antigen-binding site, and a residual “Fc” fragment, a designation reflecting the ability to crystallize readily.
  • Pepsin treatment yields an F(ab′) 2 fragment that has two antigen combining sites and is still capable of cross-linking antigen.
  • “Fv” is the minimum antibody fragment that contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the V H -V L dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • the “Fab” fragment also contains the constant domain of the light chain and the first constant domain (CH 1 ) of the heavy chain.
  • Fab fragments differ from Fab′ fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CH 1 domain including one or more cysteines from the antibody hinge region.
  • Fab′-SH is the designation herein for Fab′ in which the cysteine residue(s) of the constant domains bear a free thiol group.
  • F(ab′) 2 antibody fragments originally were produced as pairs of Fab′ fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these classes can be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA, and IgA2. The subclasses can be further divided into types, e.g., IgG2a and IgG2b.
  • Single-chain Fv or “sFv” or “scFv” antibody fragments comprise the V H and V L domains of antibody, wherein these domains are present in a single polypeptide chain.
  • the Fv polypeptide further comprises a polypeptide linker between the V H and V L domains, which enables the sFv to form the desired structure for antigen binding.
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (V H ) connected to a light-chain variable domain (V L ) in the same polypeptide chain (V H -V L ).
  • V H heavy-chain variable domain
  • V L light-chain variable domain
  • the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites.
  • Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448.
  • a “monoclonal antibody” is an antibody produced by a group of identical cells, all of which were produced from a single cell by repetitive cellular replication. That is, the clone of cells only produces a single antibody species. While a monoclonal antibody can be produced using hybridoma production technology, other production methods known to those skilled in the art can also be used (e.g., antibodies derived from antibody phage display libraries).
  • CDR complementarity determining region
  • CDRs have been described by Kabat et al., J. Biol. Chem. 252:6609-6616 (1977); Kabat et al., U.S. Dept. of Health and Human Services, “Sequences of proteins of immunological interest” (1991) (also referred to herein as Kabat 1991); by Chothia et al., J. Mol. Biol. 196:901-917 (1987) (also referred to herein as Chothia 1987); and by MacCallum et al., J. Mol. Biol.
  • CDR-L1”, CDR-L2”, and CDR-L3 refer, respectively, to the first, second, and third CDRs in a light chain variable region.
  • CDR-H1”, CDR-H2”, and CDR-H3 refer, respectively, to the first, second, and third CDRs in a heavy chain variable region.
  • CDR-1”, “CDR-2”, and “CDR-3” refer, respectively, to the first, second and third CDRs of either chain's variable region.
  • affinity refers to the equilibrium constant for the reversible binding of two agents (e.g., an antibody and an antigen) and is expressed as a dissociation constant (K D ).
  • Affinity can be at least 1-fold greater, at least 2-fold greater, at least 3-fold greater, at least 4-fold greater, at least 5-fold greater, at least 6-fold greater, at least 7-fold greater, at least 8-fold greater, at least 9-fold greater, at least 10-fold greater, at least 20-fold greater, at least 30-fold greater, at least 40-fold greater, at least 50-fold greater, at least 60-fold greater, at least 70-fold greater, at least 80-fold greater, at least 90-fold greater, at least 100-fold greater, or at least 1,000-fold greater, or more, than the affinity of an antibody for unrelated amino acid sequences.
  • Affinity of an antibody to a target protein can be, for example, from about 100 nanomolar (nM) to about 0.1 nM, from about 100 nM to about 1 picomolar (pM), or from about 100 nM to about 1 femtomolar (fM) or more.
  • nM nanomolar
  • pM picomolar
  • fM femtomolar
  • the term “avidity” refers to the resistance of a complex of two or more agents to dissociation after dilution.
  • the terms “immunoreactive” and “preferentially binds” are used interchangeably herein with respect to antibodies and/or antigen-binding fragments.
  • binding refers to a direct association between two molecules, due to, for example, covalent, electrostatic, hydrophobic, and ionic and/or hydrogen-bond interactions, including interactions such as salt bridges and water bridges.
  • An antibody of the invention binds specifically to an epitope within a LAG-3 protein, particularly a human LAG-3 protein.
  • Specific binding refers to binding with an affinity of at least about 5 ⁇ 10 ⁇ 7 M or greater, e.g., 10 ⁇ 7 M, 5 ⁇ 10 ⁇ 8 M, 10 ⁇ 8 M, or greater.
  • Non-specific binding refers to binding with an affinity of less than about 10 ⁇ 7 M, e.g., binding with an affinity of 10 ⁇ 6 M, 10 ⁇ 5 M, 10 ⁇ 4 M, etc.
  • an antibody that “specifically binds human LAG-3” is intended to refer to an antibody that binds to human LAG-3 protein (and possibly a LAG-3 protein from one or more non-human species) but does not substantially bind to non-LAG-3 proteins.
  • the antibody binds to a human LAG-3 protein with “high affinity”, namely with a of K D of 1 ⁇ 10 ⁇ 7 M or less, more preferably 1 ⁇ 10 ⁇ 8 M or less, more preferably 5 ⁇ 10 ⁇ 9 M or less, more preferably 1 ⁇ 10 ⁇ 9 M or less.
  • does not substantially bind to a protein or cells, as used herein, means does not bind or does not bind with a high affinity to the protein or cells, i.e. binds to the protein or cells with a K D of 1 ⁇ 10 ⁇ 6 M or more, more preferably 1 ⁇ 10 ⁇ 6 M or more, more preferably 1 ⁇ 10 ⁇ 4 M or more, more preferably 1 ⁇ 10 ⁇ 3 M or more, even more preferably 1 ⁇ 10 ⁇ 2 M or more.
  • K assoc or “K a ”, as used herein, is intended to refer to the association rate of a particular antibody-antigen interaction
  • K dis or “K d ,” as used herein, is intended to refer to the dissociation rate of a particular antibody-antigen interaction
  • K D is intended to refer to the dissociation constant, which is obtained from the ratio of K d to K a . (i.e., K d /K a ) and is expressed as a molar concentration (M).
  • high affinity for an IgG antibody refers to an antibody having a K D of 1 ⁇ 10 ⁇ 7 M or less, more preferably 5 ⁇ 10 ⁇ 8 M or less, even more preferably 1 ⁇ 10 ⁇ 8 M or less, even more preferably 5 ⁇ 10 ⁇ 9 M or less and even more preferably 1 ⁇ 10 ⁇ 9 M or less for a target antigen.
  • “high affinity” binding can vary for other antibody isotypes.
  • “high affinity” binding for an IgM isotype refers to an antibody having a K D of 10 ⁇ 6 M or less, more preferably 10 ⁇ 7 M or less, even more preferably 10 ⁇ 8 M or less.
  • an antibody of the invention binds to a human LAG-3 protein with higher affinity (i.e. with a lower dissociation constant) than the antagonistic anti-LAG-3 monoclonal antibody 17B4 (Baixeras, et al., J. Exp. Med., 1992, Vol. 176: 327-337).
  • Example 7 describes the results of Biacore analysis of binding of 17B4 antibody to human LAG-3Ig protein. The results show that the dissociation constant of 17B4 for human LAG-3Ig was 3.69 nM.
  • an antibody of the present invention binds a human LAG-3 protein (or a human LAG-3Ig protein) with a dissociation constant (K D ) of less than 3.69 nM, for example as determined by Biacore analysis.
  • an antibody of the present invention binds a human LAG-3 protein (or a derivative thereof, such as a human LAG-3Ig protein) with a dissociation constant (K D ) of no more than 3.5 nM, no more than 2.5 nM, no more than 2 nM, no more than 1 nM, no more than 0.9 nM, no more than 0.8 nM, no more than 0.7 nM, no more than 0.6 nM, no more than 0.5 nM, no more than 0.4 nM, no more than 0.3 nM, no more than 0.2 nM, no more than 0.1 nM.
  • K D dissociation constant
  • an anti-LAG-3 antibody of the present disclosure binds a human LAG-3 protein with a K D of no more than 90 pM, no more than 80 pM, no more than 70 pM, no more than 60 pM, no more than 50 pM, no more than 40 pM, no more than 30 pM, no more than 20 pM, no more than 10 pM, no more than 9 pM, no more than 8 pM, no more than 7 pM, no more than 6 pM, no more than 5 pM, no more than 4 pM, no more than 3 pM, no more than 2 pM, or no more than 1 pM.
  • Example 20 below describes the results of Biacore analysis of binding of chimeric 13E2 IgG4 antibody (described below), and of humanized 13E2 IgG4 (described below), to human LAG-3Ig protein.
  • the results show that the dissociation constant of chimeric 13E2 IgG4 antibody for human LAG-3Ig was 21.9 pM, and of humanized 13E2 IgG4 for human LAG-3Ig was 22.8 pM.
  • an antibody of the present invention binds a human LAG-3 protein (or a human LAG-3Ig protein) with a dissociation constant (K D ) of no more than 100 pM, no more than 90 pM, no more than 80 pM, no more than 70 pM, no more than 60 pM, no more than 50 pM, no more than 40 pM, no more than 30 pM, or no more than 25 pM, for example as determined by Biacore analysis.
  • K D dissociation constant
  • the affinity of an antibody of the invention, or antigen-binding fragment thereof may be at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% higher than the affinity of 17B4 for a human LAG-3 protein. In some embodiments, an antibody of the invention, or antigen-binding fragment thereof, may be at least 1.5, 2, 2.5, 3, or 3.5 times the affinity of 17B4 for a human LAG-3 protein.
  • the affinity of an antibody to a human LAG-3 protein can be determined by one skilled in the art, suitably by surface plasmon resonance, for example using a biosensor system, such as a Biacore system (Murphy et al, Using Biacore to measure the binding kinetics of an antibody-antigen interaction; Curr Protoc Protein Sci. 2006 September; Chapter 19: Unit 19.14).
  • Biacore analysis can be used to determine the dissociation constant between an antibody of the invention and a human LAG-3 protein.
  • Binding to human LAG-3 can be assessed using one or more other techniques also well established in the art.
  • an antibody can be tested by a flow cytometry assay in which the antibody is reacted with a cell line that expresses human LAG-3, such as CHO cells that have been transfected to express human LAG-3 on their cell surface.
  • a cell line that expresses human LAG-3 such as CHO cells that have been transfected to express human LAG-3 on their cell surface.
  • Other suitable cells for use in flow cytometry assays include SEB-stimulated PBMCs, or anti-CD3-stimulated CD4 + activated T cells, which express native LAG-3.
  • Still other suitable binding assays include ELISA assays, for example, using a recombinant LAG-3 protein.
  • An antibody of the invention is an isolated antibody.
  • An “isolated” antibody is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the antibody, and can include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes.
  • the antibody will be purified (1) to greater than 90%, greater than 95%, or greater than 98%, by weight of antibody as determined by the Lowry method, for example, more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) under reducing or non-reducing conditions using Coomassie blue or silver stain.
  • Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. In some instances, isolated antibody will be prepared by at least one purification step.
  • an antibody of the invention is a humanized antibody, or antigen-binding fragment thereof, particularly, a humanized monoclonal antibody, or antigen-binding fragment thereof.
  • a humanized antibody of the invention, or antigen-binding fragment thereof may comprise a humanized light chain framework region and/or a humanized heavy chain framework region.
  • humanized antibody refers to an immunoglobulin comprising portions of immunoglobulins of different origin, wherein at least one portion comprises amino acid sequences of human origin.
  • the humanized antibody can comprise portions derived from an immunoglobulin of nonhuman origin with the requisite specificity, such as a mouse, and from immunoglobulin sequences of human origin (e.g., immunoglobulin), joined together chemically by conventional techniques (e.g., synthetic) or prepared as a contiguous polypeptide using genetic engineering techniques (e.g., DNA encoding the protein portions of the chimeric antibody can be expressed to produce a contiguous polypeptide chain).
  • humanized immunoglobulin is an immunoglobulin containing one or more immunoglobulin chains comprising a CDR derived from an antibody of nonhuman origin and a framework region derived from a light and/or heavy chain of human origin (e.g., CDR-grafted antibodies with or without framework changes). Chimeric or CDR-grafted single chain antibodies are also encompassed by the term humanized immunoglobulin. See, e.g., Cabilly et al., U.S. Pat. No. 4,816,567; Cabilly et al., European Patent No. 0,125,023 B1; Boss et al., U.S. Pat. No.
  • variable region amino acid sequence may be identical to the variable region amino sequence of the species from which it is derived (for example, mouse sequence), or may be at least 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to that variable region sequence.
  • variable region amino acid sequence of a chimeric antibody of the invention may comprise one or more amino acid deletions, substitutions, or additions (for example one, two, three, four, five, six, seven, eight, nine, or ten amino acid deletions, substitutions, or additions) compared with the variable region amino acid sequence of the species from which it is derived.
  • the constant region amino acid sequence may be identical to the constant region amino acid sequence of the species from which it is derived (for example, human sequence), or may be at least 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to that constant region amino acid sequence.
  • the constant region amino acid sequence of a chimeric antibody of the invention may comprise one or more amino acid deletions, substitutions, or additions (for example one, two, three, four, five, six, seven, eight, nine, or ten amino acid deletions, substitutions, or additions) compared with the constant region amino acid sequence of the species from which it is derived.
  • the amino acid sequence of the Fc hinge region of a chimeric antibody may be mutated to decrease the biological half-life of the antibody, or the amino acid sequence of the Fc region may be mutated to increase the biological half-life of the chimeric antibody.
  • the heavy chain variable region sequence comprises, or is derived from a mouse antibody, and the heavy chain constant region sequence comprises, or is derived from IgG4 Fc sequence.
  • the light chain variable region sequence comprises, or is derived from a mouse antibody, and the light chain constant region sequence comprises, or is derived from human Ig kappa (IgK) chain C sequence.
  • the Fc region comprises a mutant human IgG4 Fc sequence with an S228P mutation to abolish Fab arm exchange (as shown in FIG. 20 (A) for chimeric antibody Chim13E2IgG4 comprising heavy chain sequence 13E2IgG4mut).
  • the Fc region comprises a wild-type human Ig kappa (IgK) chain C portion (13E2IgK) (as shown in FIG. 20 (B) for chimeric antibody Chim13E2IgG4 comprising light chain sequence 13E2IgK).
  • the numbering of residues in the Fc region used for the human IgG4 Fc mutant described above is the standard numbering of the Eu index as in Kabat (Kabat, E. A. et al., Sequences of proteins of immunological interest. 5th Edition—US Department of Health and Human Services, NIH publication n° 91-3242, pp 662, 680, 689 (1991)).
  • Humanized antibodies may be produced using synthetic and/or recombinant nucleic acids to prepare genes (e.g., cDNA) encoding the desired humanized chain.
  • genes e.g., cDNA
  • nucleic acid (e.g., DNA) sequences coding for humanized variable regions can be constructed using PCR mutagenesis methods to alter DNA sequences encoding a human or humanized chain, such as a DNA template from a previously humanized variable region (see e.g., Kamman, M., et al., Nucl. Acids Res., 17: 5404 (1989)); Sato, K., et al., Cancer Research, 53: 851-856 (1993); Daugherty, B. L.
  • variants can also be readily produced.
  • cloned variable regions can be mutagenized, and sequences encoding variants with the desired specificity can be selected (e.g., from a phage library; see e.g., Krebber et al., U.S. Pat. No. 5,514,548; Hoogenboom et al., WO 93/06213, published Apr. 1, 1993)).
  • variable region when used in reference to an antibody variable region is intended to mean all amino acid residues outside the CDR regions within the variable region of an antibody.
  • a variable region framework is generally a discontinuous amino acid sequence between about 100-120 amino acids in length but is intended to reference only those amino acids outside of the CDRs.
  • framework region is intended to mean each domain of the framework that is separated by the CDRs.
  • Humanization of a framework region(s) reduces the risk of the antibody eliciting a human-anti-mouse-antibody (HAMA) response in humans.
  • Art-recognized methods of determining immune response can be performed to monitor a HAMA response in a particular patient or during clinical trials. Patients administered humanized antibodies can be given an immunogenicity assessment at the beginning and throughout the administration of the therapy.
  • the HAMA response is measured, for example, by detecting antibodies to the humanized therapeutic reagent, in serum samples from the patient using a method known to one in the art, including surface plasmon resonance technology (BIACORE) and/or solid-phase enzyme-linked immunosorbent assay (ELISA) analysis.
  • BIACORE surface plasmon resonance technology
  • ELISA solid-phase enzyme-linked immunosorbent assay
  • Certain amino acids from the human variable region framework residues are selected for substitution based on their possible influence on CDR conformation and/or binding antigen.
  • the unnatural juxtaposition of murine CDR regions with human variable framework region can result in unnatural conformational restraints, which, unless corrected by substitution of certain amino acid residues, lead to loss of binding affinity.
  • the selection of amino acid residues for substitution can be determined, in part, by computer modeling.
  • Computer hardware and software for producing three-dimensional images of immunoglobulin molecules are known in the art.
  • molecular models are produced starting from solved structures for immunoglobulin chains or domains thereof.
  • the chains to be modeled are compared for amino acid sequence similarity with chains or domains of solved three-dimensional structures, and the chains or domains showing the greatest sequence similarity is/are selected as starting points for construction of the molecular model.
  • Chains or domains sharing at least 50% sequence identity are selected for modeling, e.g., those sharing at least 60%, at least 70%, at least 80%, at least 90% sequence identity or more are selected for modeling.
  • the solved starting structures are modified to allow for differences between the actual amino acids in the immunoglobulin chains or domains being modeled, and those in the starting structure.
  • the modified structures are then assembled into a composite immunoglobulin.
  • the model is refined by energy minimization and by verifying that all atoms are within appropriate distances from one another and that bond lengths and angles are within chemically acceptable limits.
  • CDR and framework regions may be as defined by Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md., 1987 and 1991).
  • An alternative structural definition has been proposed by Chothia et al., J. Mol. Biol. 196:901 (1987); Nature 342:878 (1989); and J. Mol. Biol. 186:651 (1989) (collectively referred to as “Chothia”).
  • Chothia When framework residues, as defined by Kabat, supra, constitute structural loop residues as defined by Chothia, supra, the amino acids present in the mouse antibody can be selected for substitution into the humanized antibody.
  • Residues that are “adjacent to a CDR region” include amino acid residues in positions immediately adjacent to one or more of the CDRs in the primary sequence of the humanized immunoglobulin chain, for example, in positions immediately adjacent to a CDR as defined by Kabat, or a CDR as defined by Chothia (See e.g., Chothia and Lesk J M B 196:901 (1987)). These amino acids are particularly likely to interact with the amino acids in the CDRs and, if chosen from the acceptor, to distort the donor CDRs and reduce affinity.
  • CDR and framework regions may be as defined by MacCallum et al., or Lefranc et al. (supra—see Table 1).
  • a humanized V H framework or V L framework is a consensus humanized framework.
  • a consensus humanized framework can represent the most commonly occurring amino acid residue in a selection of human immunoglobulin V L or V H framework sequences.
  • an antibody of the invention, or fragment thereof comprises one or more humanized framework regions (FRs).
  • a subject anti-LAG-3 antibody comprises a light chain variable region comprising one, two, three, or four light chain FRs that have been humanized.
  • a subject antibody comprises a light chain variable region comprising, in order from N-terminus to C-terminus: a humanized light chain FR1; a CDR-L1 as set forth herein; a humanized light chain FR2; a CDR-L2 as set forth herein; a humanized light chain FR3; a CDR-L3 as set forth herein; and a humanized light chain FR4.
  • the respective amino acid sequences of CDR-L1, CDR-L2, and CDR-L3 are: SEQ ID NO:1, SEQ ID NO:2, and SEQ ID NO:3.
  • a subject antibody can comprise a light chain variable region that comprises, in order from N-terminus to C-terminus: a humanized light chain FR1; a CDR-L1 comprising amino acid sequence SEQ ID NO:1; a humanized light chain FR2; a CDR-L2 comprising amino acid sequence SEQ ID NO:2; a humanized light chain FR3; a CDR-L3 comprising amino acid sequence SEQ ID NO:3; and a humanized light chain FR4.
  • the respective amino acid sequences of CDR-L1, CDR-L2, and CDR-L3 are: SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6.
  • a subject antibody can comprise a light chain variable region that comprises, in order from N-terminus to C-terminus: a humanized light chain FR1; a CDR-L1 comprising amino acid sequence SEQ ID NO:4; a humanized light chain FR2; a CDR-L2 comprising amino acid sequence SEQ ID NO:5; a humanized light chain FR3; a CDR-L3 comprising amino acid sequence SEQ ID NO:6; and a humanized light chain FR4.
  • the respective amino acid sequences of CDR-L1, CDR-L2, and CDR-L3 are: SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:26.
  • a subject antibody can comprise a light chain variable region that comprises, in order from N-terminus to C-terminus: a humanized light chain FR1; a CDR-L1 comprising amino acid sequence SEQ ID NO:24; a humanized light chain FR2; a CDR-L2 comprising amino acid sequence SEQ ID NO:25; a humanized light chain FR3; a CDR-L3 comprising amino acid sequence SEQ ID NO:26; and a humanized light chain FR4.
  • the respective amino acid sequences of CDR-L1, CDR-L2, and CDR-L3 are selected from: SEQ ID NO: 4 and 24 (CDR-L1); SEQ ID NO: 5 and 25 (CDR-L2); and SEQ ID NO: 6 and 26 (CDR-L3).
  • a subject antibody can comprise a light chain variable region that comprises, in order from N-terminus to C-terminus: a humanized light chain FR1; a CDR-L1 comprising amino acid sequence selected from SEQ ID NO:4 and 24; a humanized light chain FR2; a CDR-L2 comprising amino acid sequence selected from SEQ ID NO:5 and 25; a humanized light chain FR3; a CDR-L3 comprising amino acid sequence selected from SEQ ID NO:6 and 26; and a humanized light chain FR4.
  • the respective amino acid sequences of CDR-L1, CDR-L2, and CDR-L3 are: SEQ ID NO:11, SEQ ID NO:12, and SEQ ID NO:13.
  • a subject antibody can comprise a light chain variable region that comprises, in order from N-terminus to C-terminus: a humanized light chain FR1; a CDR-L1 comprising amino acid sequence SEQ ID NO:11; a humanized light chain FR2; a CDR-L2 comprising amino acid sequence SEQ ID NO:12; a humanized light chain FR3; a CDR-L3 comprising amino acid sequence SEQ ID NO:13; and a humanized light chain FR4.
  • the respective amino acid sequences of CDR-L1, CDR-L2, and CDR-L3 are: SEQ ID NO:14, SEQ ID NO:15, and SEQ ID NO:16.
  • a subject antibody can comprise a light chain variable region that comprises, in order from N-terminus to C-terminus: a humanized light chain FR1; a CDR-L1 comprising amino acid sequence SEQ ID NO:14; a humanized light chain FR2; a CDR-L2 comprising amino acid sequence SEQ ID NO:15; a humanized light chain FR3; a CDR-L3 comprising amino acid sequence SEQ ID NO:16; and a humanized light chain FR4.
  • the respective amino acid sequences of CDR-L1, CDR-L2, and CDR-L3 are: SEQ ID NO:34, SEQ ID NO:35, and SEQ ID NO:36.
  • a subject antibody can comprise a light chain variable region that comprises, in order from N-terminus to C-terminus: a humanized light chain FR1; a CDR-L1 comprising amino acid sequence SEQ ID NO:34; a humanized light chain FR2; a CDR-L2 comprising amino acid sequence SEQ ID NO:35; a humanized light chain FR3; a CDR-L3 comprising amino acid sequence SEQ ID NO:36; and a humanized light chain FR4.
  • the respective amino acid sequences of CDR-L1, CDR-L2, and CDR-L3 are selected from: SEQ ID NO: 14 and 34 (CDR-L1); SEQ ID NO: 15 and 35 (CDR-L2); and SEQ ID NO: 16 and 36 (CDR-L3).
  • a subject antibody can comprise a light chain variable region that comprises, in order from N-terminus to C-terminus: a humanized light chain FR1; a CDR-L1 comprising amino acid sequence selected from SEQ ID NO:14 and 34; a humanized light chain FR2; a CDR-L2 comprising amino acid sequence selected from SEQ ID NO:15 and 35; a humanized light chain FR3; a CDR-L3 comprising amino acid sequence selected from SEQ ID NO:16 and 36; and a humanized light chain FR4.
  • a subject anti-LAG-3 antibody comprises a heavy chain variable region comprising one, two, three, or four heavy chain FRs that have been humanized.
  • a subject antibody comprises a heavy chain variable region comprising, in order from N-terminus to C-terminus: a humanized heavy chain FR1; a CDR-H1 as set forth herein; a humanized heavy chain FR2; a CDR-H2 as set forth herein; a humanized heavy chain FR3; a CDR-H3 as set forth herein; and a humanized heavy chain FR4.
  • the respective amino acid sequences of CDR-H1, CDR-H2, and CDR-H3 are: SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6.
  • a subject antibody can comprise a heavy chain variable region that comprises, in order from N-terminus to C-terminus: a humanized heavy chain FR1; a CDR-H1 comprising amino acid sequence SEQ ID NO:4; a humanized heavy chain FR2; a CDR-H2 comprising amino acid sequence SEQ ID NO:5; a humanized heavy chain FR3; a CDR-H3 comprising amino acid sequence SEQ ID NO:6; and a humanized heavy chain FR4.
  • the respective amino acid sequences of CDR-H1, CDR-H2, and CDR-H3 are: SEQ ID NO:1, SEQ ID NO:2, and SEQ ID NO:3.
  • a subject antibody can comprise a heavy chain variable region that comprises, in order from N-terminus to C-terminus: a humanized heavy chain FR1; a CDR-H1 comprising amino acid sequence SEQ ID NO:1; a humanized heavy chain FR2; a CDR-H2 comprising amino acid sequence SEQ ID NO:2; a humanized heavy chain FR3; a CDR-H3 comprising amino acid sequence SEQ ID NO:3; and a humanized heavy chain FR4.
  • the respective amino acid sequences of CDR-H1, CDR-H2, and CDR-H3 are: SEQ ID NO:21, SEQ ID NO:22, and SEQ ID NO:23.
  • a subject antibody can comprise a heavy chain variable region that comprises, in order from N-terminus to C-terminus: a humanized heavy chain FR1; a CDR-H1 comprising amino acid sequence SEQ ID NO:21; a humanized heavy chain FR2; a CDR-H2 comprising amino acid sequence SEQ ID NO:22; a humanized heavy chain FR3; a CDR-H3 comprising amino acid sequence SEQ ID NO:23; and a humanized heavy chain FR4.
  • the respective amino acid sequences of CDR-H1, CDR-H2, and CDR-H3 are selected from: SEQ ID NO: 1 and 21 (CDR-H1); SEQ ID NO: 2 and 22 (CDR-H2); and SEQ ID NO: 3 and 23 (CDR-H3).
  • a subject antibody can comprise a heavy chain variable region that comprises, in order from N-terminus to C-terminus: a humanized heavy chain FR1; a CDR-H1 comprising amino acid sequence selected from SEQ ID NO:1 and 21; a humanized heavy chain FR2; a CDR-H2 comprising amino acid sequence selected from SEQ ID NO:2 and 22; a humanized heavy chain FR3; a CDR-H3 comprising amino acid sequence selected from SEQ ID NO:3 and 23; and a humanized heavy chain FR4.
  • the respective amino acid sequences of CDR-H1, CDR-H2, and CDR-H3 are: SEQ ID NO:14, SEQ ID NO:15, and SEQ ID NO:16.
  • a subject antibody can comprise a heavy chain variable region that comprises, in order from N-terminus to C-terminus: a humanized heavy chain FR1; a CDR-H1 comprising amino acid sequence SEQ ID NO:14; a humanized heavy chain FR2; a CDR-H2 comprising amino acid sequence SEQ ID NO:15; a humanized heavy chain FR3; a CDR-H3 comprising amino acid sequence SEQ ID NO:16; and a humanized heavy chain FR4.
  • the respective amino acid sequences of CDR-H1, CDR-H2, and CDR-H3 are: SEQ ID NO:11, SEQ ID NO:12, and SEQ ID NO:13.
  • a subject antibody can comprise a heavy chain variable region that comprises, in order from N-terminus to C-terminus: a humanized heavy chain FR1; a CDR-H1 comprising amino acid sequence SEQ ID NO:11; a humanized heavy chain FR2; a CDR-H2 comprising amino acid sequence SEQ ID NO:12; a humanized heavy chain FR3; a CDR-H3 comprising amino acid sequence SEQ ID NO:13; and a humanized heavy chain FR4.
  • the respective amino acid sequences of CDR-H1, CDR-H2, and CDR-H3 are: SEQ ID NO:31, SEQ ID NO:32, and SEQ ID NO:33.
  • a subject antibody can comprise a heavy chain variable region that comprises, in order from N-terminus to C-terminus: a humanized heavy chain FR1; a CDR-H1 comprising amino acid sequence SEQ ID NO:31; a humanized heavy chain FR2; a CDR-H2 comprising amino acid sequence SEQ ID NO:32; a humanized heavy chain FR3; a CDR-H3 comprising amino acid sequence SEQ ID NO:33; and a humanized heavy chain FR4.
  • the respective amino acid sequences of CDR-H1, CDR-H2, and CDR-H3 are selected from: SEQ ID NO: 11 and 31 (CDR-H1); SEQ ID NO: 12 and 32 (CDR-H2); and SEQ ID NO: 13 and 33 (CDR-H3).
  • a subject antibody can comprise a heavy chain variable region that comprises, in order from N-terminus to C-terminus: a humanized heavy chain FR1; a CDR-H1 comprising amino acid sequence selected from SEQ ID NO:11 and 31; a humanized heavy chain FR2; a CDR-H2 comprising amino acid sequence selected from SEQ ID NO:12 and 32; a humanized heavy chain FR3; a CDR-H3 comprising amino acid sequence selected from SEQ ID NO:13 and 33; and a humanized heavy chain FR4.
  • Suitable humanized framework sequences include:
  • VH variant 1 VH 1 ) VH 1 FR1: (SEQ ID NO: 52) QVTLKESGPALVKPTQTLTLTCTFS; VH 1 FR2: (SEQ ID NO: 53) WIRQPPGKALEWLA; VH 1 FR3: (SEQ ID NO: 54) RLTISKDTSKSQVILNMTNMDPVDTATYYC; and VH 1 FR4: (SEQ ID NO: 55) WGQGTTVTVSS; VH variant 2 (VH 2 ) VH 2 FR1: (SEQ ID NO: 56) QITLKESGPALVKPTQTLTLTCSFS; VH 2 FR2: (SEQ ID NO: 57) WIRQPPGKALEWLA; VH 2 FR3: (SEQ ID NO: 58) RLTISKDTSKNQVVLTMANMDPVDTATYYC; VH 2 FR4: (SEQ ID NO: 59) WGQGTTVTVSS; V
  • variable regions of humanized VH variants 1-4, and VL variants 1-4 are shown aligned with the corresponding sequence of the original mouse antibody 13E2 in FIG. 22 .
  • CDR sequences are highlighted in grey.
  • Changes in the humanized framework sequences of the variants, compared with the original mouse sequence, are shown underlined and in bold.
  • the changed residues in the humanized sequence for each variant are also set out in Tables 26 (heavy chain sequences) and 27 (light chain sequences) in Example 18.
  • a humanized antibody of the invention (or antigen-binding fragment thereof) comprises a humanized heavy chain which comprises any of the amino acid substitutions depicted for VH 1 , VH 2 , VH 3 , or VH 4 in Table 26 and/or a humanized light chain which comprises any of the amino acid substitutions depicted for VL 1 , VL 2 , VL 3 , or VL 4 in Table 27.
  • An antibody of the invention, or antigen-binding fragment thereof may comprise any of the above humanized framework sequences (SEQ ID NOs:52-83), or any combination of the above humanized framework sequences (SEQ ID NOs:52-83).
  • the humanized heavy chain framework region may comprise an amino acid sequence of: any of SEQ ID NOs: 52, 53, 54, or 55; any of SEQ ID NOs: 56, 57, 58, or 59; any of SEQ ID NOs: 60, 61, 62, or 63; or any of SEQ ID NOs: 64, 65, 66, or 67.
  • an antibody of the invention, or antigen-binding fragment thereof may comprise:
  • VH FR1 VH framework region 1 (VH FR1) of SEQ ID NO: 52; a VH FR2 of SEQ ID NO: 53; a VH FR3 of SEQ ID NO: 54; and a VH FR4 of SEQ ID NO: 55;
  • VH FR1 VH framework region 1 (VH FR1) of SEQ ID NO: 56; a VH FR2 of SEQ ID NO: 57; a VH FR3 of SEQ ID NO: 58; and a VH FR4 of SEQ ID NO: 59;
  • VH FR1 VH framework region 1 (VH FR1) of SEQ ID NO: 60; a VH FR2 of SEQ ID NO: 61; a VH FR3 of SEQ ID NO: 62; and a VH FR4 of SEQ ID NO: 63; or
  • VH FR1 VH framework region 1 (VH FR1) of SEQ ID NO: 64; a VH FR2 of SEQ ID NO: 65; a VH FR3 of SEQ ID NO: 66; and a VH FR4 of SEQ ID NO: 67.
  • the humanized light chain framework region may comprise an amino acid sequence of: any of SEQ ID NOs: 68, 69, 70, or 71; any of SEQ ID NOs: 72, 73, 74, or 75; any of SEQ ID NOs: 76, 77, 78, or 79; or any of SEQ ID NOs: 80, 81, 82, or 83.
  • an antibody of the invention, or antigen-binding fragment thereof may comprise:
  • VL FR1 VL framework region 1 (VL FR1) of SEQ ID NO: 68; a VL FR2 of SEQ ID NO: 69; a VL FR3 of SEQ ID NO: 70; and a VL FR4 of SEQ ID NO: 71;
  • VL FR1 VL framework region 1 (VL FR1) of SEQ ID NO: 72; a VL FR2 of SEQ ID NO: 73; a VL FR3 of SEQ ID NO: 74; and a VL FR4 of SEQ ID NO: 75;
  • VL FR1 VL framework region 1 (VL FR1) of SEQ ID NO: 76; a VL FR2 of SEQ ID NO: 77; a VL FR3 of SEQ ID NO: 78; and a VL FR4 of SEQ ID NO: 79; or
  • VL FR1 VL framework region 1 (VL FR1) of SEQ ID NO: 80; a VL FR2 of SEQ ID NO: 81; a VL FR3 of SEQ ID NO: 82; and a VL FR4 of SEQ ID NO: 83.
  • an antibody of the invention, or antigen-binding fragment thereof may comprise any of the following combinations of humanized framework sequences:
  • an antibody of the invention, or antigen-binding fragment thereof comprises the following humanized framework sequences:
  • VH 4 FR1 (SEQ ID NO: 64) QITLKESGPTLVKPTQTLTLTCTFS; VH 4 FR2: (SEQ ID NO: 65) WIRQPPGKTLEWLT; VH 4 FR3: (SEQ ID NO: 66) RLSITKDTSKNQVVLTMTNMDPLDTGTYYC; and VH 4 FR4: (SEQ ID NO: 67) WGQGTLVTVSS; and VL 3 FR1: (SEQ ID NO: 76) DIVMTQTPSSLSASVGDRVTITC; VL 3 FR2: (SEQ ID NO: 77) WYQQRPGQAPKLLIY; VL 3 FR3: (SEQ ID NO: 78) GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC; and VL 3 FR4: (SEQ ID NO: 79) FGQGTRLDIK
  • an isolated antibody of the invention, or antigen-binding fragment thereof comprises an antibody VH region comprising:
  • VH FR1 having an amino acid sequence of SEQ ID NO: 52; a VH CDR1 having an amino acid sequence selected from SEQ ID NO: 1 and 21; a VH FR2 having an amino acid sequence of SEQ ID NO: 53; a VH CDR2 having an amino acid sequence selected from SEQ ID NO: 2 and 22; a VH FR3 having an amino acid sequence of SEQ ID NO: 54; a VH CDR3 having an amino acid sequence selected from SEQ ID NO: 3 and 23; and a VH FR4 having an amino acid sequence of SEQ ID NO: 55;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 52; a VH CDR1 having an amino acid sequence of SEQ ID NO: 1; a VH FR2 having an amino acid sequence of SEQ ID NO: 53; a VH CDR2 having an amino acid sequence of SEQ ID NO: 2; a VH FR3 having an amino acid sequence of SEQ ID NO: 54; a VH CDR3 having an amino acid sequence of SEQ ID NO: 3; and a VH FR4 having an amino acid sequence of SEQ ID NO: 55;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 52; a VH CDR1 having an amino acid sequence of SEQ ID NO: 21; a VH FR2 having an amino acid sequence of SEQ ID NO: 53; a VH CDR2 having an amino acid sequence of SEQ ID NO: 22; a VH FR3 having an amino acid sequence of SEQ ID NO: 54; a VH CDR3 having an amino acid sequence of SEQ ID NO: 23; and a VH FR4 having an amino acid sequence of SEQ ID NO: 55;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 56; a VH CDR1 having an amino acid sequence selected from SEQ ID NO: 1 and 21; a VH FR2 having an amino acid sequence of SEQ ID NO: 57; a VH CDR2 having an amino acid sequence selected from SEQ ID NO: 2 and 22; a VH FR3 having an amino acid sequence of SEQ ID NO: 58; a VH CDR3 having an amino acid sequence selected from SEQ ID NO: 3 and 23; and a VH FR4 having an amino acid sequence of SEQ ID NO: 59;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 56; a VH CDR1 having an amino acid sequence of SEQ ID NO: 1; a VH FR2 having an amino acid sequence of SEQ ID NO: 57; a VH CDR2 having an amino acid sequence of SEQ ID NO: 2; a VH FR3 having an amino acid sequence of SEQ ID NO: 58; a VH CDR3 having an amino acid sequence of SEQ ID NO: 3; and a VH FR4 having an amino acid sequence of SEQ ID NO: 59;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 56; a VH CDR1 having an amino acid sequence of SEQ ID NO: 21; a VH FR2 having an amino acid sequence of SEQ ID NO: 57; a VH CDR2 having an amino acid sequence of SEQ ID NO: 22; a VH FR3 having an amino acid sequence of SEQ ID NO: 58; a VH CDR3 having an amino acid sequence of SEQ ID NO: 23; and a VH FR4 having an amino acid sequence of SEQ ID NO: 59;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 60; a VH CDR1 having an amino acid sequence selected from SEQ ID NO: 1 and 21; a VH FR2 having an amino acid sequence of SEQ ID NO: 61; a VH CDR2 having an amino acid sequence selected from SEQ ID NO: 2 and 22; a VH FR3 having an amino acid sequence of SEQ ID NO: 62; a VH CDR3 having an amino acid sequence selected from SEQ ID NO: 3 and 23; and a VH FR4 having an amino acid sequence of SEQ ID NO: 63;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 60; a VH CDR1 having an amino acid sequence of SEQ ID NO: 1; a VH FR2 having an amino acid sequence of SEQ ID NO: 61; a VH CDR2 having an amino acid sequence of SEQ ID NO: 2; a VH FR3 having an amino acid sequence of SEQ ID NO: 62; a VH CDR3 having an amino acid sequence of SEQ ID NO: 3; and a VH FR4 having an amino acid sequence of SEQ ID NO: 63;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 60; a VH CDR1 having an amino acid sequence of SEQ ID NO: 21; a VH FR2 having an amino acid sequence of SEQ ID NO: 61; a VH CDR2 having an amino acid sequence of SEQ ID NO: 22; a VH FR3 having an amino acid sequence of SEQ ID NO: 62; a VH CDR3 having an amino acid sequence of SEQ ID NO: 23; and a VH FR4 having an amino acid sequence of SEQ ID NO: 63;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 64; a VH CDR1 having an amino acid sequence selected from SEQ ID NO: 1 and 21; a VH FR2 having an amino acid sequence of SEQ ID NO: 65; a VH CDR2 having an amino acid sequence selected from SEQ ID NO: 2 and 22; a VH FR3 having an amino acid sequence of SEQ ID NO: 66; a VH CDR3 having an amino acid sequence selected from SEQ ID NO: 3 and 23; and a VH FR4 having an amino acid sequence of SEQ ID NO: 67;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 64; a VH CDR1 having an amino acid sequence of SEQ ID NO: 1; a VH FR2 having an amino acid sequence of SEQ ID NO: 65; a VH CDR2 having an amino acid sequence of SEQ ID NO: 2; a VH FR3 having an amino acid sequence of SEQ ID NO: 66; a VH CDR3 having an amino acid sequence of SEQ ID NO: 3; and a VH FR4 having an amino acid sequence of SEQ ID NO: 67; or
  • VH FR1 having an amino acid sequence of SEQ ID NO: 64; a VH CDR1 having an amino acid sequence of SEQ ID NO: 21; a VH FR2 having an amino acid sequence of SEQ ID NO: 65; a VH CDR2 having an amino acid sequence of SEQ ID NO: 22; a VH FR3 having an amino acid sequence of SEQ ID NO: 66; a VH CDR3 having an amino acid sequence of SEQ ID NO: 23; and a VH FR4 having an amino acid sequence of SEQ ID NO: 67.
  • an isolated antibody of the invention comprises an antibody heavy chain comprising amino acid sequence of SEQ ID NO:84.
  • an isolated antibody of the invention, or antigen-binding fragment thereof comprises an antibody VL region comprising:
  • VL FR1 having an amino acid sequence of SEQ ID NO: 68; a VL CDR1 having an amino acid sequence selected from SEQ ID NO: 4 and 24; a VL FR2 having an amino acid sequence of SEQ ID NO: 69; a VL CDR2 having an amino acid sequence selected from SEQ ID NO: 5 and 25; a VL FR3 having an amino acid sequence of SEQ ID NO: 70; a VL CDR3 having an amino acid sequence selected from SEQ ID NO: 6 and 26; and a VL FR4 having an amino acid sequence of SEQ ID NO: 71;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 68; a VL CDR1 having an amino acid sequence of SEQ ID NO: 4; a VL FR2 having an amino acid sequence of SEQ ID NO: 69; a VL CDR2 having an amino acid sequence of SEQ ID NO: 5; a VL FR3 having an amino acid sequence of SEQ ID NO: 70; a VL CDR3 having an amino acid sequence of SEQ ID NO: 6; and a VL FR4 having an amino acid sequence of SEQ ID NO: 71;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 68; a VL CDR1 having an amino acid sequence of SEQ ID NO: 24; a VL FR2 having an amino acid sequence of SEQ ID NO: 69; a VL CDR2 having an amino acid sequence of SEQ ID NO: 25; a VL FR3 having an amino acid sequence of SEQ ID NO: 70; a VL CDR3 having an amino acid sequence of SEQ ID NO: 26; and a VL FR4 having an amino acid sequence of SEQ ID NO: 71;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 72; a VL CDR1 having an amino acid sequence selected from SEQ ID NO: 4 and 24; a VL FR2 having an amino acid sequence of SEQ ID NO: 73; a VL CDR2 having an amino acid sequence selected from SEQ ID NO: 5 and 25; a VL FR3 having an amino acid sequence of SEQ ID NO: 74; a VL CDR3 having an amino acid sequence selected from SEQ ID NO: 6 and 26; and a VL FR4 having an amino acid sequence of SEQ ID NO: 75;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 72; a VL CDR1 having an amino acid sequence of SEQ ID NO: 4; a VL FR2 having an amino acid sequence of SEQ ID NO: 73; a VL CDR2 having an amino acid sequence of SEQ ID NO: 5; a VL FR3 having an amino acid sequence of SEQ ID NO: 74; a VL CDR3 having an amino acid sequence of SEQ ID NO: 6; and a VL FR4 having an amino acid sequence of SEQ ID NO: 75;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 72; a VL CDR1 having an amino acid sequence of SEQ ID NO: 24; a VL FR2 having an amino acid sequence of SEQ ID NO: 73; a VL CDR2 having an amino acid sequence of SEQ ID NO: 25; a VL FR3 having an amino acid sequence of SEQ ID NO: 74; a VL CDR3 having an amino acid sequence of SEQ ID NO: 26; and a VL FR4 having an amino acid sequence of SEQ ID NO: 75;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 76; a VL CDR1 having an amino acid sequence selected from SEQ ID NO: 4 and 24; a VL FR2 having an amino acid sequence of SEQ ID NO: 77; a VL CDR2 having an amino acid sequence selected from SEQ ID NO: 5 and 25; a VL FR3 having an amino acid sequence of SEQ ID NO: 78; a VL CDR3 having an amino acid sequence selected from SEQ ID NO: 6 and 26; and a VL FR4 having an amino acid sequence of SEQ ID NO: 79;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 76; a VL CDR1 having an amino acid sequence of SEQ ID NO: 4; a VL FR2 having an amino acid sequence of SEQ ID NO: 77; a VL CDR2 having an amino acid sequence of SEQ ID NO: 5; a VL FR3 having an amino acid sequence of SEQ ID NO: 78; a VL CDR3 having an amino acid sequence of SEQ ID NO: 6; and a VL FR4 having an amino acid sequence of SEQ ID NO: 79;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 76; a VL CDR1 having an amino acid sequence of SEQ ID NO: 24; a VL FR2 having an amino acid sequence of SEQ ID NO: 77; a VL CDR2 having an amino acid sequence of SEQ ID NO: 25; a VL FR3 having an amino acid sequence of SEQ ID NO: 78; a VL CDR3 having an amino acid sequence of SEQ ID NO: 26; and a VL FR4 having an amino acid sequence of SEQ ID NO: 79;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 80; a VL CDR1 having an amino acid sequence selected from SEQ ID NO: 4 and 24; a VL FR2 having an amino acid sequence of SEQ ID NO: 81; a VL CDR2 having an amino acid sequence selected from SEQ ID NO: 5 and 25; a VL FR3 having an amino acid sequence of SEQ ID NO: 82; a VL CDR3 having an amino acid sequence selected from SEQ ID NO: 6 and 26; and a VL FR4 having an amino acid sequence of SEQ ID NO: 83;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 80; a VL CDR1 having an amino acid sequence of SEQ ID NO: 4; a VL FR2 having an amino acid sequence of SEQ ID NO: 81; a VL CDR2 having an amino acid sequence of SEQ ID NO: 5; a VL FR3 having an amino acid sequence of SEQ ID NO: 82; a VL CDR3 having an amino acid sequence of SEQ ID NO: 6; and a VL FR4 having an amino acid sequence of SEQ ID NO: 83; or
  • VL FR1 having an amino acid sequence of SEQ ID NO: 80; a VL CDR1 having an amino acid sequence of SEQ ID NO: 24; a VL FR2 having an amino acid sequence of SEQ ID NO: 81; a VL CDR2 having an amino acid sequence of SEQ ID NO: 25; a VL FR3 having an amino acid sequence of SEQ ID NO: 82; a VL CDR3 having an amino acid sequence of SEQ ID NO: 26; and a VL FR4 having an amino acid sequence of SEQ ID NO: 83.
  • an isolated antibody of the invention comprises an antibody light chain comprising amino acid sequence of SEQ ID NO:85.
  • an isolated antibody of the invention comprises an antibody heavy chain comprising amino acid sequence of SEQ ID NO:84, and an antibody light chain comprising amino acid sequence of SEQ ID NO:85.
  • an isolated antibody of the invention, or antigen-binding fragment thereof comprises an antibody VH region comprising:
  • VH FR1 having an amino acid sequence of SEQ ID NO: 52; a VH CDR1 having an amino acid sequence selected from SEQ ID NO: 11 and 31; a VH FR2 having an amino acid sequence of SEQ ID NO: 53; a VH CDR2 having an amino acid sequence selected from SEQ ID NO: 12 and 32; a VH FR3 having an amino acid sequence of SEQ ID NO: 54; a VH CDR3 having an amino acid sequence selected from SEQ ID NO: 13 and 33; and a VH FR4 having an amino acid sequence of SEQ ID NO: 55;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 52; a VH CDR1 having an amino acid sequence of SEQ ID NO: 11; a VH FR2 having an amino acid sequence of SEQ ID NO: 53; a VH CDR2 having an amino acid sequence of SEQ ID NO: 12; a VH FR3 having an amino acid sequence of SEQ ID NO: 54; a VH CDR3 having an amino acid sequence of SEQ ID NO: 13; and a VH FR4 having an amino acid sequence of SEQ ID NO: 55;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 52; a VH CDR1 having an amino acid sequence of SEQ ID NO: 31; a VH FR2 having an amino acid sequence of SEQ ID NO: 53; a VH CDR2 having an amino acid sequence of SEQ ID NO: 32; a VH FR3 having an amino acid sequence of SEQ ID NO: 54; a VH CDR3 having an amino acid sequence of SEQ ID NO: 33; and a VH FR4 having an amino acid sequence of SEQ ID NO: 55;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 56; a VH CDR1 having an amino acid sequence selected from SEQ ID NO: 11 and 31; a VH FR2 having an amino acid sequence of SEQ ID NO: 57; a VH CDR2 having an amino acid sequence selected from SEQ ID NO: 12 and 32; a VH FR3 having an amino acid sequence of SEQ ID NO: 58; a VH CDR3 having an amino acid sequence selected from SEQ ID NO: 13 and 33; and a VH FR4 having an amino acid sequence of SEQ ID NO: 59;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 56; a VH CDR1 having an amino acid sequence of SEQ ID NO: 11; a VH FR2 having an amino acid sequence of SEQ ID NO: 57; a VH CDR2 having an amino acid sequence of SEQ ID NO: 12; a VH FR3 having an amino acid sequence of SEQ ID NO: 58; a VH CDR3 having an amino acid sequence of SEQ ID NO: 13; and a VH FR4 having an amino acid sequence of SEQ ID NO: 59;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 56; a VH CDR1 having an amino acid sequence of SEQ ID NO: 31; a VH FR2 having an amino acid sequence of SEQ ID NO: 57; a VH CDR2 having an amino acid sequence of SEQ ID NO: 32; a VH FR3 having an amino acid sequence of SEQ ID NO: 58; a VH CDR3 having an amino acid sequence of SEQ ID NO: 33; and a VH FR4 having an amino acid sequence of SEQ ID NO: 59;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 60; a VH CDR1 having an amino acid sequence selected from SEQ ID NO: 11 and 31; a VH FR2 having an amino acid sequence of SEQ ID NO: 61; a VH CDR2 having an amino acid sequence selected from SEQ ID NO: 12 and 32; a VH FR3 having an amino acid sequence of SEQ ID NO: 62; a VH CDR3 having an amino acid sequence selected from SEQ ID NO: 13 and 33; and a VH FR4 having an amino acid sequence of SEQ ID NO: 63;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 60; a VH CDR1 having an amino acid sequence of SEQ ID NO: 11; a VH FR2 having an amino acid sequence of SEQ ID NO: 61; a VH CDR2 having an amino acid sequence of SEQ ID NO: 12; a VH FR3 having an amino acid sequence of SEQ ID NO: 62; a VH CDR3 having an amino acid sequence of SEQ ID NO: 13; and a VH FR4 having an amino acid sequence of SEQ ID NO: 63;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 60; a VH CDR1 having an amino acid sequence of SEQ ID NO: 31; a VH FR2 having an amino acid sequence of SEQ ID NO: 61; a VH CDR2 having an amino acid sequence of SEQ ID NO: 32; a VH FR3 having an amino acid sequence of SEQ ID NO: 62; a VH CDR3 having an amino acid sequence of SEQ ID NO: 33; and a VH FR4 having an amino acid sequence of SEQ ID NO: 63;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 64; a VH CDR1 having an amino acid sequence selected from SEQ ID NO: 11 and 31; a VH FR2 having an amino acid sequence of SEQ ID NO: 65; a VH CDR2 having an amino acid sequence selected from SEQ ID NO: 12 and 32; a VH FR3 having an amino acid sequence of SEQ ID NO: 66; a VH CDR3 having an amino acid sequence selected from SEQ ID NO: 13 and 33; and a VH FR4 having an amino acid sequence of SEQ ID NO: 67;
  • VH FR1 having an amino acid sequence of SEQ ID NO: 64; a VH CDR1 having an amino acid sequence of SEQ ID NO: 11; a VH FR2 having an amino acid sequence of SEQ ID NO: 65; a VH CDR2 having an amino acid sequence of SEQ ID NO: 12; a VH FR3 having an amino acid sequence of SEQ ID NO: 66; a VH CDR3 having an amino acid sequence of SEQ ID NO: 13; and a VH FR4 having an amino acid sequence of SEQ ID NO: 67; or
  • VH FR1 having an amino acid sequence of SEQ ID NO: 64; a VH CDR1 having an amino acid sequence of SEQ ID NO: 31; a VH FR2 having an amino acid sequence of SEQ ID NO: 65; a VH CDR2 having an amino acid sequence of SEQ ID NO: 32; a VH FR3 having an amino acid sequence of SEQ ID NO: 66; a VH CDR3 having an amino acid sequence of SEQ ID NO: 33; and a VH FR4 having an amino acid sequence of SEQ ID NO: 67.
  • an isolated antibody of the invention, or antigen-binding fragment thereof comprises an antibody VL region comprising:
  • VL FR1 having an amino acid sequence of SEQ ID NO: 68; a VL CDR1 having an amino acid sequence selected from SEQ ID NO: 14 and 34; a VL FR2 having an amino acid sequence of SEQ ID NO: 69; a VL CDR2 having an amino acid sequence selected from SEQ ID NO: 15 and 35; a VL FR3 having an amino acid sequence of SEQ ID NO: 70; a VL CDR3 having an amino acid sequence selected from SEQ ID NO: 16 and 36; and a VL FR4 having an amino acid sequence of SEQ ID NO: 71;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 68; a VL CDR1 having an amino acid sequence of SEQ ID NO: 14; a VL FR2 having an amino acid sequence of SEQ ID NO: 69; a VL CDR2 having an amino acid sequence of SEQ ID NO: 15; a VL FR3 having an amino acid sequence of SEQ ID NO: 70; a VL CDR3 having an amino acid sequence of SEQ ID NO: 16; and a VL FR4 having an amino acid sequence of SEQ ID NO: 71;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 68; a VL CDR1 having an amino acid sequence of SEQ ID NO: 34; a VL FR2 having an amino acid sequence of SEQ ID NO: 69; a VL CDR2 having an amino acid sequence of SEQ ID NO: 35; a VL FR3 having an amino acid sequence of SEQ ID NO: 70; a VL CDR3 having an amino acid sequence of SEQ ID NO: 36; and a VL FR4 having an amino acid sequence of SEQ ID NO: 71;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 72; a VL CDR1 having an amino acid sequence selected from SEQ ID NO: 14 and 34; a VL FR2 having an amino acid sequence of SEQ ID NO: 73; a VL CDR2 having an amino acid sequence selected from SEQ ID NO: 15 and 35; a VL FR3 having an amino acid sequence of SEQ ID NO: 74; a VL CDR3 having an amino acid sequence selected from SEQ ID NO: 16 and 36; and a VL FR4 having an amino acid sequence of SEQ ID NO: 75;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 72; a VL CDR1 having an amino acid sequence of SEQ ID NO: 14; a VL FR2 having an amino acid sequence of SEQ ID NO: 73; a VL CDR2 having an amino acid sequence of SEQ ID NO: 15; a VL FR3 having an amino acid sequence of SEQ ID NO: 74; a VL CDR3 having an amino acid sequence of SEQ ID NO: 16; and a VL FR4 having an amino acid sequence of SEQ ID NO: 75;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 72; a VL CDR1 having an amino acid sequence of SEQ ID NO: 34; a VL FR2 having an amino acid sequence of SEQ ID NO: 73; a VL CDR2 having an amino acid sequence of SEQ ID NO: 35; a VL FR3 having an amino acid sequence of SEQ ID NO: 74; a VL CDR3 having an amino acid sequence of SEQ ID NO: 36; and a VL FR4 having an amino acid sequence of SEQ ID NO: 75;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 76; a VL CDR1 having an amino acid sequence selected from SEQ ID NO: 14 and 34; a VL FR2 having an amino acid sequence of SEQ ID NO: 77; a VL CDR2 having an amino acid sequence selected from SEQ ID NO: 15 and 35; a VL FR3 having an amino acid sequence of SEQ ID NO: 78; a VL CDR3 having an amino acid sequence selected from SEQ ID NO: 16 and 36; and a VL FR4 having an amino acid sequence of SEQ ID NO: 79;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 76; a VL CDR1 having an amino acid sequence of SEQ ID NO: 14; a VL FR2 having an amino acid sequence of SEQ ID NO: 77; a VL CDR2 having an amino acid sequence of SEQ ID NO: 15; a VL FR3 having an amino acid sequence of SEQ ID NO: 78; a VL CDR3 having an amino acid sequence of SEQ ID NO: 16; and a VL FR4 having an amino acid sequence of SEQ ID NO: 79;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 76; a VL CDR1 having an amino acid sequence of SEQ ID NO: 34; a VL FR2 having an amino acid sequence of SEQ ID NO: 77; a VL CDR2 having an amino acid sequence of SEQ ID NO: 35; a VL FR3 having an amino acid sequence of SEQ ID NO: 78; a VL CDR3 having an amino acid sequence of SEQ ID NO: 36; and a VL FR4 having an amino acid sequence of SEQ ID NO: 79;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 80; a VL CDR1 having an amino acid sequence selected from SEQ ID NO: 14 and 34; a VL FR2 having an amino acid sequence of SEQ ID NO: 81; a VL CDR2 having an amino acid sequence selected from SEQ ID NO: 15 and 35; a VL FR3 having an amino acid sequence of SEQ ID NO: 82; a VL CDR3 having an amino acid sequence selected from SEQ ID NO: 16 and 36; and a VL FR4 having an amino acid sequence of SEQ ID NO: 83;
  • VL FR1 having an amino acid sequence of SEQ ID NO: 80; a VL CDR1 having an amino acid sequence of SEQ ID NO: 14; a VL FR2 having an amino acid sequence of SEQ ID NO: 81; a VL CDR2 having an amino acid sequence of SEQ ID NO: 15; a VL FR3 having an amino acid sequence of SEQ ID NO: 82; a VL CDR3 having an amino acid sequence of SEQ ID NO: 16; and a VL FR4 having an amino acid sequence of SEQ ID NO: 83; or
  • a subject antibody comprises a constant region of an immunoglobulin (e.g., an Fc region).
  • the Fc region if present, is a human Fc region. If constant regions are present, the antibody can contain both light chain and heavy chain constant regions. Suitable heavy chain constant regions include CH1, hinge, CH2, CH3, and CH4 regions.
  • a suitable heavy chain Fc region is a human isotype IgG4 Fc.
  • Light chain constant regions can be lambda or kappa.
  • a subject antibody e.g., a subject humanized antibody
  • a subject antibody can comprise sequences from more than one class or isotype.
  • Antibodies can be expressed as tetramers containing two light and two heavy chains, as separate heavy chains, light chains, as Fab, Fab′, F(ab′) 2 , and Fv, or as single chain antibodies in which heavy and light chain variable domains are linked through a spacer.
  • the heavy chain region is of the isotype IgG4.
  • the hinge region comprises an S241P substitution. See, e.g., Angal et al. (1993) Mol. Immunol. 30:105.
  • the hinge region comprises an L236E (or L235E, using EU numbering; Kabat et al. (1991) Sequences of Proteins of Immunological Interest, 5 th Ed. U.S. Dept. Health and Human Services, Bethesda, Md., NIH Publication No. 91-3242) substitution. See, e.g., Reddy et al. (2000) J. Immunol. 164:1925; and Klechevsky et al. (2010) Blood 116:1685.
  • the hinge region comprises an S241P substitution and an L236E substitution.
  • the constant region amino acid sequence may be identical to the constant region amino acid sequence of the species from which it is derived (for example, human sequence), or may be at least 60%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to that constant region amino acid sequence.
  • the constant region amino acid sequence of an antibody of the invention may comprise one or more amino acid deletions, substitutions, or additions (for example one, two, three, four, five, six, seven, eight, nine, or ten amino acid deletions, substitutions, or additions) compared with the constant region amino acid sequence of the species from which it is derived.
  • amino acid sequence of the Fc hinge region of an antibody of the invention may be mutated to decrease the biological half-life of the antibody, or the amino acid sequence of the Fc region may be mutated to increase the biological half-life of the antibody.
  • an antibody of the invention or antigen-binding fragment thereof, lacks antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) so that the antibody, or fragment, can be used to negatively regulate T cell proliferation and/or function without depleting T cells as a result of ADCC or CDC.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • the Fc region comprises a wild-type human IgG4 Fc sequence.
  • the Fc region comprises a mutant human IgG4 Fc sequence with an S228P mutation to abolish Fab arm exchange (as shown in FIG. 20 (A) for chimeric antibody Chim13E2IgG4 comprising heavy chain sequence 13E2IgG4mut).
  • the Fc region comprises a wild-type human Ig kappa (IgK) chain C portion (13E2IgK) (as shown in FIG. 20 (B) for chimeric antibody Chim13E2IgG4 comprising light chain sequence 13E2IgK).
  • the numbering of residues in the Fc region used for the human IgG4 Fc mutant described above is the standard numbering of the EU index as in Kabat (Kabat, E. A. et al., Sequences of proteins of immunological interest. 5th Edition—US Department of Health and Human Services, NIH publication n° 91-3242, pp 662, 680, 689 (1991)).
  • a subject antibody can comprise a free thiol (—SH) group at the carboxyl terminus, where the free thiol group can be used to attach the antibody to a second polypeptide (e.g., another antibody, including a subject antibody), a scaffold, a carrier, etc.
  • a second polypeptide e.g., another antibody, including a subject antibody
  • a subject antibody comprises one or more non-naturally occurring amino acids.
  • the non-naturally encoded amino acid comprises a carbonyl group, an acetyl group, an aminooxy group, a hydrazine group, a hydrazide group, a semicarbazide group, an azide group, or an alkyne group. See, e.g., U.S. Pat. No. 7,632,924 for suitable non-naturally occurring amino acids. Inclusion of a non-naturally occurring amino acid can provide for linkage to a polymer, a second polypeptide, a scaffold, etc.
  • a subject antibody linked to a water-soluble polymer can be made by reacting a water-soluble polymer (e.g., PEG) that comprises a carbonyl group to the antibody, where the antibody comprises a non-naturally encoded amino acid that comprises an aminooxy, hydrazine, hydrazide or semicarbazide group.
  • a subject antibody linked to a water-soluble polymer can be made by reacting a subject antibody that comprises an alkyne-containing amino acid with a water-soluble polymer (e.g., PEG) that comprises an azide moiety; in some embodiments, the azide or alkyne group is linked to the PEG molecule through an amide linkage.
  • non-naturally encoded amino acid refers to an amino acid that is not one of the 20 common amino acids or pyrrolysine or selenocysteine.
  • Other terms that can be used synonymously with the term “non-naturally encoded amino acid” are “non-natural amino acid,” “unnatural amino acid,” “non-naturally-occurring amino acid,” and variously hyphenated and non-hyphenated versions thereof.
  • the term “non-naturally encoded amino acid” also includes, but is not limited to, amino acids that occur by modification (e.g.
  • a naturally encoded amino acid including but not limited to, the 20 common amino acids or pyrrolysine and selenocysteine
  • non-naturally-occurring amino acids include, but are not limited to, N-acetylglucosaminyl-L-serine, N-acetylglucosaminyl-L-threonine, and O-phosphotyrosine.
  • a subject antibody is linked (e.g., covalently linked) to a polymer (e.g., a polymer other than a polypeptide).
  • a polymer e.g., a polymer other than a polypeptide.
  • Suitable polymers include, e.g., biocompatible polymers, and water-soluble biocompatible polymers.
  • Suitable polymers include synthetic polymers and naturally-occurring polymers.
  • Suitable polymers include, e.g., substituted or unsubstituted straight or branched chain polyalkylene, polyalkenylene or polyoxyalkylene polymers or branched or unbranched polysaccharides, e.g. a homo- or hetero-polysaccharide.
  • Suitable polymers include, e.g., ethylene vinyl alcohol copolymer (commonly known by the generic name EVOH or by the trade name EVAL); polybutylmethacrylate; poly(hydroxyvalerate); poly(L-lactic acid); polycaprolactone; poly(lactide-co-glycolide); poly(hydroxybutyrate); poly(hydroxybutyrate-co-valerate); polydioxanone; polyorthoester; polyanhydride; poly(glycolic acid); poly(D,L-lactic acid); poly(glycolic acid-co-trimethylene carbonate); polyphosphoester; polyphosphoester urethane; poly(amino acids); cyanoacrylates; poly(trimethylene carbonate); poly(iminocarbonate); copoly(ether-esters) (e.g., poly(ethylene oxide)-poly(lactic acid) (PEO/PLA) co-polymers); polyalkylene oxalates; polyphosphazene
  • Suitable synthetic polymers include unsubstituted and substituted straight or branched chain poly(ethyleneglycol), poly(propyleneglycol) poly(vinylalcohol), and derivatives thereof, e.g., substituted poly(ethyleneglycol) such as methoxypoly(ethyleneglycol), and derivatives thereof.
  • Suitable naturally-occurring polymers include, e.g., albumin, amylose, dextran, glycogen, and derivatives thereof.
  • Suitable polymers can have an average molecular weight in a range of from 500 Da to 50,000 Da, e.g., from 5,000 Da to 40,000 Da, or from 25,000 to 40,000 Da.
  • a subject antibody comprises a poly(ethylene glycol) (PEG) or methoxypoly(ethyleneglycol) polymer
  • the PEG or methoxypoly(ethyleneglycol) polymer can have a molecular weight in a range of from about 0.5 kiloDaltons (kDa) to 1 kDa, from about 1 kDa to 5 kDa, from 5 kDa to 10 kDa, from 10 kDa to 25 kDa, from 25 kDa to 40 kDa, or from 40 kDa to 60 kDa.
  • kDa kiloDaltons
  • a subject antibody is covalently linked to a non-peptide synthetic polymer.
  • a subject antibody is covalently linked to a PEG polymer.
  • a subject scFv multimer is covalently linked to a PEG polymer. See, e.g., Albrecht et al. (2006) J. Immunol. Methods 310:100. Methods and reagents suitable for PEGylation of a protein are well known in the art and can be found in, e.g., U.S. Pat. No. 5,849,860.
  • PEG suitable for conjugation to a protein is generally soluble in water at room temperature, and has the general formula R(O—CH 2 —CH 2 ) n O—R, where R is hydrogen or a protective group such as an alkyl or an alkanol group, and where n is an integer from 1 to 1,000. Where R is a protective group, it generally has from 1 to 8 carbons.
  • the PEG conjugated to the subject antibody is linear. In some embodiments, the PEG conjugated to the subject antibody is branched. Branched PEG derivatives such as those described in U.S. Pat. No. 5,643,575, “star-PEG's” and multi-armed PEG's such as those described in Shearwater Polymers, Inc. catalog “Polyethylene Glycol Derivatives 1997-1998.” Star PEGs are described in the art including, e.g., in U.S. Pat. No. 6,046,305.
  • a subject antibody can be glycosylated, e.g., a subject antibody can comprise a covalently linked carbohydrate or polysaccharide moiety.
  • Glycosylation of antibodies is typically either N-linked or O-linked.
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site.
  • O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine can also be used.
  • glycosylation sites to an antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • the alteration can also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original antibody (for O-linked glycosylation sites).
  • removal of glycosylation sites can be accomplished by amino acid alteration within the native glycosylation sites of an antibody.
  • a subject antibody will in some embodiments comprise a “radiopaque” label, e.g. a label that can be easily visualized using for example x-rays.
  • Radiopaque materials are well known to those of skill in the art. The most common radiopaque materials include iodide, bromide or barium salts. Other radiopaque materials are also known and include, but are not limited to organic bismuth derivatives (see, e.g., U.S. Pat. No. 5,939,045), radiopaque multiurethanes (see U.S. Pat. No. 5,346,981), organobismuth composites (see, e.g., U.S. Pat. No. 5,256,334), radiopaque barium multimer complexes (see, e.g., U.S. Pat. No. 4,866,132), and the like.
  • a subject antibody can be covalently linked to a second moiety (e.g., a lipid, a polypeptide other than a subject antibody, a synthetic polymer, a carbohydrate, and the like) using for example, glutaraldehyde, a homobifunctional cross-linker, or a heterobifunctional cross-linker.
  • Glutaraldehyde cross-links polypeptides via their amino moieties.
  • Homobifunctional cross-linkers e.g., a homobifunctional imidoester, a homobifunctional N-hydroxysuccinimidyl (NHS) ester, or a homobifunctional sulfhydryl reactive cross-linker
  • a homobifunctional imidoester e.g., a homobifunctional N-hydroxysuccinimidyl (NHS) ester, or a homobifunctional sulfhydryl reactive cross-linker
  • a homobifunctional imidoester e.g., a homobifunctional N-hydroxysuccinimidyl (NHS) ester, or a homobifunctional sulfhydryl reactive cross-linker
  • Homobifunctional NHS ester and imido esters cross-link amine containing polypeptides. In a mild alkaline pH, imido esters react only with primary amines to form imidoamides, and overall charge of the cross-linked
  • Homobifunctional sulfhydryl reactive cross-linkers include bismaleimidhexane (BMH), 1,5-difluoro-2,4-dinitrobenzene (DFDNB), and 1,4-di-(3′,2′-pyridyldithio) propinoamido butane (DPDPB).
  • BMH bismaleimidhexane
  • DFDNB 1,5-difluoro-2,4-dinitrobenzene
  • DPDPB 1,4-di-(3′,2′-pyridyldithio) propinoamido butane
  • Heterobifunctional cross-linkers have two or more different reactive moieties (e.g., amine reactive moiety and a sulfhydryl-reactive moiety) and are cross-linked with one of the polypeptides via the amine or sulfhydryl reactive moiety, then reacted with the other polypeptide via the non-reacted moiety.
  • Multiple heterobifunctional haloacetyl cross-linkers are available, as are pyridyl disulfide cross-linkers.
  • Carbodiimides are a classic example of heterobifunctional cross-linking reagents for coupling carboxyls to amines, which results in an amide bond.
  • a subject antibody can be immobilized on a solid support.
  • Suitable supports are well known in the art and comprise, inter alia, commercially available column materials, polystyrene beads, latex beads, magnetic beads, colloid metal particles, glass and/or silicon chips and surfaces, nitrocellulose strips, nylon membranes, sheets, duracytes, wells of reaction trays (e.g., multi-well plates), plastic tubes, etc.
  • a solid support can comprise any of a variety of substances, including, e.g., glass, polystyrene, polyvinyl chloride, polypropylene, polyethylene, polycarbonate, dextran, nylon, amylose, natural and modified celluloses, polyacrylamides, agaroses, and magnetite.
  • Solid supports can be soluble or insoluble, e.g., in aqueous solution.
  • a suitable solid support is generally insoluble in an aqueous solution.
  • a subject antibody will in some embodiments comprise a detectable label.
  • Suitable detectable labels include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means.
  • Suitable labels include, but are not limited to, magnetic beads (e.g.
  • DynabeadsTM fluorescent dyes (e.g., fluorescein isothiocyanate, texas red, rhodamine, a green fluorescent protein, a red fluorescent protein, a yellow fluorescent protein, and the like), radiolabels (e.g., 3 H, 125 I, 35 S, 14 C, or 32 P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase, luciferase, and others commonly used in an enzyme-linked immunosorbent assay (ELISA)), and colorimetric labels such as colloidal gold or colored glass or plastic (e.g. polystyrene, polypropylene, latex, etc.) beads.
  • fluorescent dyes e.g., fluorescein isothiocyanate, texas red, rhodamine, a green fluorescent protein, a red fluorescent protein, a yellow fluorescent protein, and the like
  • radiolabels e.g., 3 H, 125
  • a subject antibody comprises a contrast agent or a radioisotope, where the contrast agent or radioisotope is one that is suitable for use in imaging, e.g., imaging procedures carried out on humans.
  • labels include radioisotope such as 1231 I (iodine), 18 F (fluorine), 99 Tc (technetium), 111 In (indium), and 67 Ga (gallium), and contrast agent such as gadolinium (Gd), dysprosium, and iron.
  • Radioactive Gd isotopes ( 153 Gd) also are available and suitable for imaging procedures in non-human mammals.
  • a subject antibody can be labeled using standard techniques.
  • a subject antibody can be iodinated using chloramine T or 1,3,4,6-tetrachloro-3 ⁇ ,6 ⁇ -diphenylglycouril.
  • fluorination fluorine is added to a subject antibody during the synthesis by a fluoride ion displacement reaction. See, Muller-Gartner, H., TIB Tech., 16:122-130 (1998) and Saji, H., Crit. Rev. Ther. Drug Carrier Syst., 16(2):209-244 (1999) for a review of synthesis of proteins with such radioisotopes.
  • a subject antibody can also be labeled with a contrast agent through standard techniques.
  • a subject antibody can be labeled with Gd by conjugating low molecular Gd chelates such as Gd diethylene triamine pentaacetic acid (GdDTPA) or Gd tetraazacyclododecanetetraacetic (GdDOTA) to the antibody.
  • GdDTPA Gd diethylene triamine pentaacetic acid
  • GdDOTA Gd tetraazacyclododecanetetraacetic
  • a subject antibody can be labeled with Gd by, for example, conjugating polylysine-Gd chelates to the antibody. See, for example, Curtet et al., Invest. Radiol., 33(10):752-761 (1998).
  • a subject antibody can be labeled with Gd by incubating paramagnetic polymerized liposomes that include Gd chelator lipid with avidin and biotinylated antibody. See, for example, Sipkins et al., Nature Med., 4:623-626 (1998).
  • Suitable fluorescent proteins that can be linked to a subject antibody include, but are not limited to, a green fluorescent protein from Aequoria victoria or a mutant or derivative thereof e.g., as described in U.S. Pat. Nos. 6,066,476; 6,020,192; 5,985,577; 5,976,796; 5,968,750; 5,968,738; 5,958,713; 5,919,445; 5,874,304; e.g., Enhanced GFP, many such GFP which are available commercially, e.g., from Clontech, Inc.; a red fluorescent protein; a yellow fluorescent protein; any of a variety of fluorescent and colored proteins from Anthozoan species, as described in, e.g., Matz et al. (1999) Nature Biotechnol. 17:969-973; and the like.
  • a subject antibody is conjugated to a therapeutic. Any of the subject antibodies disclosed herein can be used to form an antibody-agent conjugate.
  • the agent can be attached to the N terminus of the light chain, the C terminus of the light chain, the N terminus of the heavy chain, or the C terminus of the heavy chain. In some embodiments, the agent is attached to the hinge of the antibody or to one or more other sites on the antibody. For a single chain antibody, the agent can be attached to the N or C terminus of the single chain antibody.
  • the agent can be conjugated to the antibody directly or via a linker using techniques known to those skilled in the art.
  • the linker can be cleavable or non-cleavable. Examples of such therapeutic agents (e.g., for use in therapy) are known to those skilled in the art.
  • a subject antibody will in some embodiments be linked to (e.g., covalently or non-covalently linked) a fusion partner, e.g., a ligand; an epitope tag; a peptide; a protein other than an antibody; and the like.
  • a fusion partner e.g., a ligand; an epitope tag; a peptide; a protein other than an antibody; and the like.
  • Suitable fusion partners include peptides and polypeptides that confer enhanced stability in vivo (e.g., enhanced serum half-life); provide ease of purification, e.g., (His) n , e.g., 6His, and the like; provide for secretion of the fusion protein from a cell; provide an epitope tag, e.g., GST, hemagglutinin (HA; e.g., YPYDVPDYA; SEQ ID NO:41), FLAG (e.g., DYKDDDDK; SEQ ID NO:42), c-myc (e.g., EQKLISEEDL; SEQ ID NO:43), and the like; provide a detectable signal, e.g., an enzyme that generates a detectable product (e.g., ⁇ -galactosidase, luciferase), or a protein that is itself detectable, e.g., a green fluorescent protein, a red fluorescent
  • the fusion can also include an affinity domain, including peptide sequences that can interact with a binding partner, e.g., such as one immobilized on a solid support, useful for identification or purification.
  • a binding partner e.g., such as one immobilized on a solid support
  • Consecutive single amino acids, such as histidine when fused to a protein, can be used for one-step purification of the fusion protein by high affinity binding to a resin column, such as nickel sepharose.
  • affinity domains include His5 (HHHHH) (SEQ ID NO:44), HisX6 (HHHHHH) (SEQ ID NO:45), c-myc (EQKLISEEDL) (SEQ ID NO:46), Flag (DYKDDDDK) (SEQ ID NO:42), StrepTag (WSHPQFEK) (SEQ ID NO:47), hemagglutinin, e.g., HA Tag (YPYDVPDYA; SEQ ID NO:41), glutathinone-S-transferase (GST), thioredoxin, cellulose binding domain, RYIRS (SEQ ID NO:48), Phe-His-His-Thr (SEQ ID NO:49), chitin binding domain, S-peptide, T7 peptide, SH2 domain, C-end RNA tag, WEAAAREACCRECCARA (SEQ ID NO:50), metal binding domains, e.g., zinc binding domains or calcium binding domains such as those from calcium-binding proteins
  • nucleotide and amino acid sequences the term “identical” or “identity” indicates the degree of identity between two nucleic acid or two amino acid sequences when optimally aligned and compared with appropriate insertions or deletions.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described below.
  • Percent identity between a query nucleic acid sequence and a subject nucleic acid sequence is the “Identities” value, expressed as a percentage, which is calculated by the BLASTN algorithm when a subject nucleic acid sequence has 100% query coverage with a query nucleic acid sequence after a pair-wise BLASTN alignment is performed.
  • Such pair-wise BLASTN alignments between a query nucleic acid sequence and a subject nucleic acid sequence are performed by using the default settings of the BLASTN algorithm available on the National Center for Biotechnology Institute's website with the filter for low complexity regions turned off.
  • a query nucleic acid sequence may be described by a nucleic acid sequence identified in one or more claims herein.
  • Percent identity between a query amino acid sequence and a subject amino acid sequence is the “Identities” value, expressed as a percentage, which is calculated by the BLASTP algorithm when a subject amino acid sequence has 100% query coverage with a query amino acid sequence after a pair-wise BLASTP alignment is performed.
  • Such pair-wise BLASTP alignments between a query amino acid sequence and a subject amino acid sequence are performed by using the default settings of the BLASTP algorithm available on the National Center for Biotechnology Institute's website with the filter for low complexity regions turned off.
  • a query amino acid sequence may be described by an amino acid sequence identified in one or more claims herein.
  • a subject antibody can be produced by any known method, e.g., conventional synthetic methods for protein synthesis; recombinant DNA methods; etc.
  • the subject antibody is produced by a method selected from the group consisting of recombinant production and chemical synthesis.
  • a subject antibody is a single chain polypeptide
  • it can be synthesized using standard chemical peptide synthesis techniques.
  • the synthesis can proceed via liquid-phase or solid-phase.
  • Solid phase polypeptide synthesis SPPS
  • Fmoc and Boc Various forms of SPPS, such as Fmoc and Boc, are available for synthesizing a subject antibody.
  • Techniques for solid phase synthesis are described by Barany and Merrifield, Solid-Phase Peptide Synthesis; pp.
  • the free N-terminal amine of a solid-phase attached is coupled to a single N-protected amino acid unit. This unit is then deprotected, revealing a new N-terminal amine to which a further amino acid can be attached.
  • the peptide remains immobilized on the solid-phase and undergoes a filtration process before being cleaved off.
  • Standard recombinant methods can be used for production of a subject antibody.
  • nucleic acids encoding light and heavy chain variable regions, optionally linked to constant regions are inserted into expression vectors.
  • the light and heavy chains can be cloned in the same or different expression vectors.
  • the DNA segments encoding immunoglobulin chains are operably linked to control sequences in the expression vector(s) that ensure the expression of immunoglobulin polypeptides.
  • Expression control sequences include, but are not limited to, promoters (e.g., naturally-associated or heterologous promoters), signal sequences, enhancer elements, repressor elements, and transcription termination sequences.
  • the expression control sequences can be eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells (e.g., COS or CHO cells). Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and the collection and purification of the antibodies.
  • eukaryotic host cells e.g., COS or CHO cells.
  • nucleic acid sequences can encode each immunoglobulin amino acid sequence.
  • the desired nucleic acid sequences can be produced by de novo solid-phase DNA synthesis or by polymerase chain reaction (PCR) mutagenesis of an earlier prepared variant of the desired polynucleotide.
  • Oligonucleotide-mediated mutagenesis is an example of a suitable method for preparing substitution, deletion and insertion variants of target polypeptide DNA. See Adelman et al., DNA 2:183 (1983). Briefly, the target polypeptide DNA is altered by hybridizing an oligonucleotide encoding the desired mutation to a single-stranded DNA template. After hybridization, a DNA polymerase is used to synthesize an entire second complementary strand of the template that incorporates the oligonucleotide primer, and encodes the selected alteration in the target polypeptide DNA.
  • Suitable expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA. Commonly, expression vectors contain selection markers (e.g., ampicillin-resistance, hygromycin-resistance, tetracycline resistance, kanamycin resistance or neomycin resistance) to permit detection of those cells transformed with the desired DNA sequences.
  • selection markers e.g., ampicillin-resistance, hygromycin-resistance, tetracycline resistance, kanamycin resistance or neomycin resistance
  • Escherichia coli is an example of a prokaryotic host cell that can be used for cloning a subject antibody-encoding polynucleotide.
  • Other microbial hosts suitable for use include bacilli, such as Bacillus subtilis , and other enterobacteriaceae, such as Salmonella, Serratia , and various Pseudomonas species.
  • bacilli such as Bacillus subtilis
  • enterobacteriaceae such as Salmonella, Serratia
  • various Pseudomonas species include Salmonella, Serratia , and various Pseudomonas species.
  • expression vectors which will typically contain expression control sequences compatible with the host cell (e.g., an origin of replication).
  • any number of a variety of well-known promoters will be present, such as the lactose promoter system, a tryptophan (trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda.
  • the promoters will typically control expression, optionally with an operator sequence, and have ribosome binding site sequences and the like, for initiating and completing transcription and translation.
  • yeast Other microbes, such as yeast, are also useful for expression.
  • Saccharomyces e.g., S. cerevisiae
  • Pichia are examples of suitable yeast host cells, with suitable vectors having expression control sequences (e.g., promoters), an origin of replication, termination sequences and the like as desired.
  • Typical promoters include 3-phosphoglycerate kinase and other glycolytic enzymes.
  • Inducible yeast promoters include, among others, promoters from alcohol dehydrogenase, isocytochrome C, and enzymes responsible for maltose and galactose utilization.
  • mammalian cells e.g., mammalian cells grown in in vitro cell culture
  • an anti-LAG-3 antibody of the present disclosure e.g., polynucleotides encoding a subject anti-LAG-3 antibody.
  • Suitable mammalian host cells include CHO cell lines, various Cos cell lines, HeLa cells, myeloma cell lines, and transformed B-cells or hybridomas.
  • Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, and an enhancer (Queen et al., Immunol. Rev. 89:49 (1986)), and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences.
  • suitable expression control sequences are promoters derived from immunoglobulin genes, SV40, adenovirus, bovine papilloma virus, cytomegalovirus and the like. See Co et al., J. Immunol. 148:1149 (1992).
  • antibodies of the invention can be produced in mice (see, for example, Laffleur et al “Production of human or humanized antibodies in mice”, Methods Mol Biol. 2012; 901:149-59).
  • the whole antibodies, their dimers, individual light and heavy chains, or other forms of a subject antibody can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, high performance liquid chromatography (HPLC) purification, gel electrophoresis, and the like (see generally Scopes, Protein Purification (Springer-Verlag, N.Y., (1982)).
  • a subject antibody can be substantially pure, e.g., at least about 80% to 85% pure, at least about 85% to 90% pure, at least about 90% to 95% pure, or 98% to 99%, or more, pure, e.g., free from contaminants such as cell debris, macromolecules other than a subject antibody, etc.
  • the present invention also provides nucleic acid molecules comprising nucleotide sequences encoding an anti-LAG-3 antibody of the invention, or antigen-binding fragment thereof.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a heavy chain variable region that is at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identical to the amino acid sequence set forth in SEQ ID NO:7 or SEQ ID NO:17.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a heavy chain variable region comprising the amino acid sequence set forth in SEQ ID NO:7 or SEQ ID NO:17.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a light chain variable region that is at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% amino acid sequence identical to the amino acid sequence set forth in SEQ ID NO:8 or SEQ ID NO:18.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a light chain variable region comprising the amino acid sequence set forth in SEQ ID NO:8 or SEQ ID NO:18.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a heavy chain variable region comprising a CDR-H1, a CDR-H2, and a CDR-H3 of SEQ ID NO:1, SEQ ID NO:2, and SEQ ID NO:3, respectively.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a light chain variable region comprising a CDR-L1, a CDR-L2, and a CDR-L3 of SEQ ID NO:4, SEQ ID NO:5, and SEQ ID NO:6, respectively.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a heavy chain variable region comprising a CDR-H1, a CDR-H2, and a CDR-H3 of SEQ ID NO:11, SEQ ID NO:12, and SEQ ID NO:13, respectively.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a light chain variable region comprising a CDR-L1, a CDR-L2, and a CDR-L3 of SEQ ID NO:14, SEQ ID NO:15, and SEQ ID NO:16, respectively.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a heavy chain variable region comprising a CDR-H1, a CDR-H2, and a CDR-H3 of SEQ ID NO:21, SEQ ID NO:22, and SEQ ID NO:23, respectively.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a light chain variable region comprising a CDR-L1, a CDR-L2, and a CDR-L3 of SEQ ID NO:24, SEQ ID NO:25, and SEQ ID NO:26, respectively.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a heavy chain variable region comprising a CDR-H1, a CDR-H2, and a CDR-H3 of SEQ ID NO:31, SEQ ID NO:32, and SEQ ID NO:33, respectively.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a light chain variable region comprising a CDR-L1, a CDR-L2, and a CDR-L3 of SEQ ID NO:34, SEQ ID NO:35, and SEQ ID NO:36, respectively.
  • a nucleic acid molecule of the present disclosure encodes a subject anti-LAG-3 antibody comprising a light chain variable region and a heavy chain variable region.
  • a nucleic acid molecule encoding a subject antibody can be operably linked to one or more regulatory elements, such as a promoter and enhancer, that allow expression of the nucleotide sequence in the intended target cells (e.g., a cell that is genetically modified to synthesize the encoded antibody).
  • regulatory elements such as a promoter and enhancer
  • Suitable promoter and enhancer elements are known in the art.
  • Suitable promoters for use in prokaryotic host cells include, but are not limited to, a bacteriophage T7 RNA polymerase promoter; a T3 promoter; a T5 promoter; a lambda P promoter; a trp promoter; a lac operon promoter; a hybrid promoter, e.g., a lac/tac hybrid promoter, a tac/trc hybrid promoter, a trp/lac promoter, a T7/lac promoter; a trc promoter; a tac promoter, and the like; a gpt promoter; an araBAD promoter; in vivo regulated promoters, such as an ssaG promoter or a related promoter (see, e.g., U.S.
  • Patent Publication No. 20040131637 discloses a pagC promoter (Pulkkinen and Miller, J. Bacteriol., 1991: 173(1): 86-93; Alpuche-Aranda et al., PNAS, 1992; 89(21): 10079-83), a nirB promoter (Harborne et al. (1992) Mol. Micro. 6:2805-2813), and the like (see, e.g., Dunstan et al. (1999) Infect. Immun. 67:5133-5141; McKelvie et al. (2004) Vaccine 22:3243-3255; and Chatfield et al. (1992) Biotechnol.
  • sigma70 promoter e.g., a consensus sigma70 promoter (see, e.g., GenBank Accession Nos. AX798980, AX798961, and AX798183); a stationary phase promoter, e.g., a dps promoter, an spv promoter, and the like; a promoter derived from the pathogenicity island SPI-2 (see, e.g., WO96/17951); an actA promoter (see, e.g., Shetron-Rama et al. (2002) Infect. Immun.
  • Suitable strong promoters for use in prokaryotes such as Escherichia coli include, but are not limited to Trc, Tac, T5, T7, and P Lambda .
  • operators for use in bacterial host cells include a lactose promoter operator (LacI repressor protein changes conformation when contacted with lactose, thereby preventing the LacI repressor protein from binding the operator), a tryptophan promoter operator (when complexed with tryptophan, TrpR repressor protein has a conformation that binds the operator; in the absence of tryptophan, the TrpR repressor protein has a conformation that does not bind the operator), and a tac promoter operator (see, for example, deBoer et al. (1983) Proc. Natl. Acad. Sci. U.S.A. 80:21-25).
  • a suitable promoter is a constitutive promoter such as an ADH1 promoter, a PGK1 promoter, an ENO promoter, a PYK1 promoter and the like; or a regulatable promoter such as a GAL1 promoter, a GAL10 promoter, an ADH2 promoter, a PHOS promoter, a CUP1 promoter, a GAL7 promoter, a MET25 promoter, a MET3 promoter, a CYC1 promoter, a HIS3 promoter, an ADH1 promoter, a PGK promoter, a GAPDH promoter, an ADC1 promoter, a TRP1 promoter, a URA3 promoter, a LEU2 promoter, an ENO promoter, a TP1 promoter, and AOX1 (e.g., for use in Pichia ).
  • a constitutive promoter such as an ADH1 promoter, a PGK1 promoter, an ENO promote
  • suitable promoters include, but are not limited to, light and/or heavy chain immunoglobulin gene promoter and enhancer elements; cytomegalovirus immediate early promoter; herpes simplex virus thymidine kinase promoter; early and late SV40 promoters; promoter present in long terminal repeats from a retrovirus; mouse metallothionein-I promoter; and various art-known tissue specific promoters.
  • a nucleic acid molecule encoding a subject antibody can be present in an expression vector and/or a cloning vector.
  • the present disclosure provides a recombinant vector, which comprises a nucleic acid molecule encoding a subject antibody in a cloning vector.
  • the present disclosure also provides a recombinant molecule, which comprises a nucleic acid molecule encoding a subject antibody operatively linked to appropriate regulatory sequence(s) in an expression vector to ensure expression of the encoded antibody.
  • nucleic acid molecules encoding the two polypeptides can be cloned in the same or separate vectors to form one or more recombinant molecules.
  • a recombinant molecule can include a selectable marker, an origin of replication, and other features that provide for replication and/or maintenance of the recombinant molecule.
  • Bacterial pBs, phagescript, PsiX174, pBluescript SK, pBs KS, pNH8a, pNH16a, pNH18a, pNH46a (Stratagene, La Jolla, Calif., USA); pTrc99A, pKK223-3, pKK233-3, pDR540, and pRIT5 (Pharmacia, Uppsala, Sweden).
  • Eukaryotic pWLneo, pSV2cat, pOG44, PXR1, pSG (Stratagene) pSVK3, pBPV, pMSG and pSVL (Pharmacia).
  • Expression vectors generally have convenient restriction sites located near the promoter sequence to provide for the insertion of nucleic acid sequences encoding heterologous proteins.
  • a selectable marker operative in the expression host can be present.
  • Suitable expression vectors include, but are not limited to, viral vectors.
  • viral vectors include, but are not limited to, viral vectors based on: vaccinia virus; poliovirus; adenovirus (see, e.g., Li et al., Invest Opthalmol Vis Sci 35:2543 2549, 1994; Borras et al., Gene Ther 6:515 524, 1999; Li and Davidson, PNAS 92:7700 7704, 1995; Sakamoto et al., H Gene Ther 5:1088 1097, 1999; WO 94/12649, WO 93/03769; WO 93/19191; WO 94/28938; WO 95/11984 and WO 95/00655); adeno-associated virus (see, e.g., Ali et al., Hum Gene Ther 9:81 86, 1998, Flannery et al., PNAS 94:6916 6921, 1997; Bennett et al., Invest Opthalmol Vis Sci 38:2857 2863,
  • SV40 herpes simplex virus
  • a retroviral vector e.g., Murine Leukemia Virus, spleen necrosis virus, and vectors derived from retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, human immunodeficiency virus (see, e.g., Miyoshi et al., PNAS 94:10319 23, 1997; Takahashi et al., J Virol 73:7812 7816, 1999), myeloproliferative sarcoma virus, and mammary tumor virus); and the like.
  • a retroviral vector e.g., Murine Leukemia Virus, spleen necrosis virus, and vectors derived from retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, human immunodeficiency virus (see, e.g., Miyoshi et al.,
  • a subject nucleic acid molecule comprises a nucleotide sequence encoding an anti-LAG-3 antibody of the present disclosure.
  • a subject nucleic acid molecule comprises a nucleotide sequence encoding heavy- and light-chain CDRs of a subject 13E2 or 34F4 antibody.
  • a subject nucleic acid molecule comprises a nucleotide sequence encoding heavy- and light-chain CDRs of a subject antibody, where the CDR-encoding sequences are interspersed with FR-encoding nucleotide sequences.
  • the FR-encoding nucleotide sequences are human FR-encoding nucleotide sequences.
  • the present disclosure provides isolated genetically modified host cells (e.g., in vitro cells) that are genetically modified with a subject nucleic acid molecule.
  • a subject isolated genetically modified host cell can produce a subject antibody.
  • Such a cell is referred to as a recombinant cell.
  • a recombinant cell comprises a recombinant molecule encoding a subject antibody.
  • Suitable host cells include eukaryotic host cells, such as a mammalian cell, an insect host cell, a yeast cell; and prokaryotic cells, such as a bacterial cell.
  • eukaryotic host cells such as a mammalian cell, an insect host cell, a yeast cell
  • prokaryotic cells such as a bacterial cell.
  • Introduction of a subject nucleic acid into the host cell can be effected, for example by calcium phosphate precipitation, DEAE dextran mediated transfection, liposome-mediated transfection, electroporation, or other known method.
  • Suitable mammalian cells include primary cells and immortalized cell lines.
  • Suitable mammalian cell lines include human cell lines, non-human primate cell lines, rodent (e.g., mouse, rat) cell lines, and the like.
  • Suitable mammalian cell lines include, but are not limited to, HeLa cells (e.g., American Type Culture Collection (ATCC) No. CCL-2), CHO cells (e.g., ATCC Nos. CRL9618, CCL61, CRL9096), 293 cells (e.g., ATCC No. CRL-1573), Vero cells, NIH 3T3 cells (e.g., ATCC No. CRL-1658), Huh-7 cells, BHK cells (e.g., ATCC No.
  • ATCC American Type Culture Collection
  • the cells are HEK cells.
  • the cells are CHO cells, e.g., CHO-K1 cells (ATCC No. CCL-61), CHO-M cells, CHO-DG44 cells (ATCC No. PTA-3356), and the like.
  • the host cell is a COS cell. In some embodiments, the host cell is a 293 cell. In some embodiments, the host cell is a CHO cell.
  • Suitable yeast cells include, but are not limited to, Pichia pastoris, Pichia finlandica, Pichia trehalophila, Pichia koclamae, Pichia membranaefaciens, Pichia opuntiae, Pichia thermotolerans, Pichia salictaria, Pichia guercuum, Pichia pijperi, Pichia stiptis, Pichia methanolica, Pichia sp., Saccharomyces cerevisiae, Saccharomyces sp., Hansenula polymorpha, Kluyveromyces sp., Kluyveromyces lactis, Candida albicans, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Trichoderma reesei, Chrysosporium lucknowense, Fusarium sp., Fusarium gramineum, Fusarium
  • Suitable prokaryotic cells include, but are not limited to, any of a variety of laboratory strains of Escherichia coli, Bacillus (e.g., B. subtilis ), Lactobacillus sp., and the like. See, e.g., Carrier et al. (1992) J. Immunol. 148:1176-1181; U.S. Pat. No. 6,447,784; and Sizemore et al. (1995) Science 270:299-302.
  • the laboratory strain is one that is non-pathogenic.
  • the host cell is Escherichia coli .
  • the host cell is Bacillus subtilis.
  • a subject antibody composition can comprise, in addition to a subject antibody, one or more of: a salt, e.g., NaCl, MgCl 2 , KCl, MgSO 4 , etc.; a buffering agent, e.g., a phosphate buffer, a citrate buffer, a Tris buffer, N-(2-Hydroxyethyl)piperazine-N′-(2-ethanesulfonic acid) (HEPES), 2-(N-Morpholino)ethanesulfonic acid (MES), 2-(N-Morpholino)ethanesulfonic acid sodium salt (MES), 3-(N-Morpholino)propanesulfonic acid (MOPS), N-tris[Hydroxymethyl]methyl-3-am inopropanesulfonic acid (TAPS), etc.; a solubilizing agent; a detergent, e.g., a phosphate buffer, a citrate buffer,
  • compositions including pharmaceutical compositions comprising a subject antibody.
  • a pharmaceutical composition also referred to herein as a formulation, comprises an effective amount of a subject antibody.
  • An “effective amount” means a dosage sufficient to produce a desired result, e.g., reduction in an adverse symptom associated with an immune disorder, amelioration of a symptom of an immune disorder, slowing progression of an immune disorder, etc.
  • the desired result is at least a reduction in a symptom of an immune disorder, as compared to a control.
  • a subject antibody can be administered to the host using any convenient means capable of resulting in the desired therapeutic effect or diagnostic effect.
  • the agent can be incorporated into a variety of formulations for therapeutic administration.
  • a subject antibody can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers, pharmaceutically acceptable diluents, or other pharmaceutically acceptable excipients and can be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.
  • a pharmaceutical composition comprises a subject antibody and a pharmaceutically acceptable excipient.
  • a subject antibody in pharmaceutical dosage forms, can be administered in the form of their pharmaceutically acceptable salts, or they can also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds.
  • the following methods and excipients are merely exemplary and are in no way limiting.
  • a subject antibody can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
  • disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
  • lubricants such as talc or magnesium stearate
  • a subject antibody can be formulated into preparations for injection by dissolving, suspending or emulsifying the antibody in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, propylene glycol, synthetic aliphatic acid glycerides, injectable organic esters (e.g., ethyl oleate), esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's, or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's dextrose), and the like.
  • the pharmaceutical composition of the present disclosure can comprise further agents such as dopamine or psychopharmacologic drugs, depending on the intended use of the pharmaceutical composition.
  • compositions comprising a subject antibody are prepared by mixing a subject antibody having the desired degree of purity with optional physiologically acceptable carriers, other excipients, stabilizers, surfactants, buffers and/or tonicity agents.
  • Acceptable carriers, other excipients and/or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid, glutathione, cysteine, methionine and citric acid; preservatives (such as ethanol, benzyl alcohol, phenol, m-cresol, p-chlor-m-cresol, methyl or propyl parabens, benzalkonium chloride, or combinations thereof); amino acids such as arginine, glycine, ornithine, lysine, histidine, glutamic acid, aspartic acid, isoleucine, leucine, alanine, phenylalanine, tyrosine,
  • the pharmaceutical composition can be in a liquid form, a lyophilized form or a liquid form reconstituted from a lyophilized form, wherein the lyophilized preparation is to be reconstituted with a sterile solution prior to administration.
  • the standard procedure for reconstituting a lyophilized composition is to add back a volume of pure water (typically equivalent to the volume removed during lyophilization); however solutions comprising antibacterial agents can be used for the production of pharmaceutical compositions for parenteral administration; see also Chen (1992) Drug Dev Ind Pharm 18, 1311-54.
  • Exemplary antibody concentrations in a subject pharmaceutical composition can range from about 1 mg/mL to about 200 mg/mL or from about 50 mg/mL to about 200 mg/mL, or from about 150 mg/mL to about 200 mg/mL.
  • An aqueous formulation of the antibody can be prepared in a pH-buffered solution, e.g., at pH ranging from about 4.0 to about 7.0, or from about 5.0 to about 6.0, or alternatively about 5.5.
  • buffers that are suitable for a pH within this range include phosphate-, histidine-, citrate-, succinate-, acetate-buffers and other organic acid buffers.
  • the buffer concentration can be from about 1 mM to about 100 mM, or from about 5 mM to about 50 mM, depending, e.g., on the buffer and the desired tonicity of the formulation.
  • a tonicity agent can be included in the antibody formulation to modulate the tonicity of the formulation.
  • exemplary tonicity agents include sodium chloride, potassium chloride, glycerin and any component from the group of amino acids, sugars as well as combinations thereof.
  • the aqueous formulation is isotonic, although hypertonic or hypotonic solutions can be suitable.
  • isotonic denotes a solution having the same tonicity as some other solution with which it is compared, such as a physiological salt solution or serum.
  • Tonicity agents can be used in an amount of about 5 mM to about 350 mM, e.g., in an amount of 100 mM to 350 nM.
  • a surfactant can also be added to the antibody formulation to reduce aggregation of the formulated antibody and/or minimize the formation of particulates in the formulation and/or reduce adsorption.
  • exemplary surfactants include polyoxyethylensorbitan fatty acid esters (Tween), polyoxyethylene alkyl ethers (Brij), alkylphenylpolyoxyethylene ethers (Triton-X), polyoxyethylene-polyoxypropylene copolymer (Poloxamer, Pluronic), and sodium dodecyl sulfate (SDS).
  • Suitable polyoxyethylenesorbitan-fatty acid esters are polysorbate 20, (sold under the trademark Tween20TM) and polysorbate 80 (sold under the trademark Tween 80TM).
  • suitable polyethylene-polypropylene copolymers are those sold under the names Pluronic® F68 or Poloxamer 188TM.
  • suitable Polyoxyethylene alkyl ethers are those sold under the trademark BrijTM.
  • Exemplary concentrations of surfactant can range from about 0.001% to about 1% w/v.
  • a lyoprotectant can also be added in order to protect the labile active ingredient (e.g. a protein) against destabilizing conditions during the lyophilization process.
  • lyoprotectants include sugars (including glucose and sucrose); polyols (including mannitol, sorbitol and glycerol); and amino acids (including alanine, glycine and glutamic acid). Lyoprotectants can be included in an amount of about 10 mM to 500 nM.
  • a subject formulation includes a subject antibody, and one or more of the above-identified agents (e.g., a surfactant, a buffer, a stabilizer, a tonicity agent) and is essentially free of one or more preservatives, such as ethanol, benzyl alcohol, phenol, m-cresol, p-chlor-m-cresol, methyl or propyl parabens, benzalkonium chloride, and combinations thereof.
  • a preservative is included in the formulation, e.g., at concentrations ranging from about 0.001 to about 2% (w/v).
  • a subject formulation can be a liquid or lyophilized formulation suitable for parenteral administration, and can comprise: about 1 mg/mL to about 200 mg/mL of a subject antibody; about 0.001% to about 1% of at least one surfactant; about 1 mM to about 100 mM of a buffer; optionally about 10 mM to about 500 mM of a stabilizer; and about 5 mM to about 305 mM of a tonicity agent; and has a pH of about 4.0 to about 7.0.
  • a subject parenteral formulation is a liquid or lyophilized formulation comprising: about 1 mg/mL to about 200 mg/mL of a subject antibody; 0.04% Tween 20 w/v; 20 mM L-histidine; and 250 mM Sucrose; and has a pH of 5.5.
  • a subject parenteral formulation comprises a lyophilized formulation comprising: 1) 15 mg/mL of a subject antibody; 0.04% Tween 20 w/v; 20 mM L-histidine; and 250 mM sucrose; and has a pH of 5.5; or 2) 75 mg/mL of a subject antibody; 0.04% Tween 20 w/v; 20 mM L-histidine; and 250 mM sucrose; and has a pH of 5.5; or 3) 75 mg/mL of a subject antibody; 0.02% Tween 20 w/v; 20 mM L-histidine; and 250 mM sucrose; and has a pH of 5.5; or 4) 75 mg/mL of a subject antibody; 0.04% Tween 20 w/v; 20 mM L-histidine; and 250 mM trehalose; and has a pH of 5.5; or 5) 75 mg/mL of a subject antibody; 0.02% Tween 20 w/v
  • a subject parenteral formulation is a liquid formulation comprising: 1) 7.5 mg/mL of a subject antibody; 0.02% Tween 20 w/v; 120 mM L-histidine; and 250 125 mM sucrose; and has a pH of 5.5; or 2) 37.5 mg/mL of a subject antibody; 0.02% Tween 20 w/v; 10 mM L-histidine; and 125 mM sucrose; and has a pH of 5.5; or 3) 37.5 mg/mL of a subject antibody; 0.01% Tween 20 w/v; 10 mM L-histidine; and 125 mM sucrose; and has a pH of 5.5; or 4) 37.5 mg/mL of a subject antibody; 0.02% Tween 20 w/v; 10 mM L-histidine; 125 mM trehalose; and has a pH of 5.5; or 5) 37.5 mg/mL of a subject antibody; 0.0
  • a subject antibody can be utilized in aerosol formulation to be administered via inhalation.
  • a subject antibody can be formulated into pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen and the like.
  • Aerosol formulations such as nasal spray formulations include purified aqueous or other solutions of the active agent with preservative agents and isotonic agents. Such formulations are adjusted to a pH and isotonic state compatible with the nasal mucous membranes.
  • a subject antibody can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases.
  • bases such as emulsifying bases or water-soluble bases.
  • a subject antibody can be administered rectally via a suppository.
  • the suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at room temperature.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions can be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition.
  • unit dosage forms for injection or intravenous administration can comprise a subject antibody in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of an anti-LAG-3 antibody of the present disclosure, calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for a subject antibody can depend on the particular antibody employed and the effect to be achieved, and the pharmacodynamics associated with each antibody in the host.
  • a subject antibody can be formulated in suppositories and, in some cases, aerosol and intranasal compositions.
  • the vehicle composition will include traditional binders and carriers such as, polyalkylene glycols, or triglycerides.
  • suppositories can be formed from mixtures containing the active ingredient in the range of about 0.5% to about 10% (w/w), e.g., about 1% to about 2%.
  • Intranasal formulations will usually include vehicles that neither cause irritation to the nasal mucosa nor significantly disturb ciliary function. Diluents such as water, aqueous saline or other known substances can be employed.
  • the nasal formulations can also contain preservatives such as, but not limited to, chlorobutanol and benzalkonium chloride.
  • a surfactant can be present to enhance absorption of the subject antibody by the nasal mucosa.
  • a subject antibody can be administered as an injectable formulation.
  • injectable compositions are prepared as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection can also be prepared.
  • the preparation can also be emulsified or the antibody encapsulated in liposome vehicles.
  • Suitable excipient vehicles are, for example, water, saline, dextrose, glycerol, ethanol, or the like, and combinations thereof.
  • the vehicle can contain minor amounts of auxiliary substances such as wetting or emulsifying agents or pH buffering agents.
  • auxiliary substances such as wetting or emulsifying agents or pH buffering agents.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., 17th edition, 1985.
  • the composition or formulation to be administered will, in any event, contain a quantity of a subject antibody adequate to achieve the desired state in the subject being treated.
  • the pharmaceutically acceptable excipients such as vehicles, adjuvants, carriers or diluents, are readily available to the public.
  • pharmaceutically acceptable auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
  • a subject antibody is formulated in a controlled release formulation.
  • Sustained-release preparations can be prepared using methods well known in the art. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody in which the matrices are in the form of shaped articles, e.g. films or microcapsules. Examples of sustained-release matrices include polyesters, copolymers of L-glutamic acid and ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, hydrogels, polylactides, degradable lactic acid-glycolic acid copolymers and poly-D-( ⁇ )-3-hydroxybutyric acid. Possible loss of biological activity and possible changes in immunogenicity of antibodies comprised in sustained-release preparations can be prevented by using appropriate additives, by controlling moisture content and by developing specific polymer matrix compositions.
  • Controlled release within the scope of the present disclosure can be taken to mean any one of a number of extended release dosage forms.
  • the following terms can be considered to be substantially equivalent to controlled release, for the purposes of the present disclosure: continuous release, controlled release, delayed release, depot, extended release, gradual release, immediate release, long-term release, programmed release, prolonged release, proportionate release, protracted release, repository, retard, slow release, spaced release, sustained release, time coat, timed release, delayed action, extended action, layered-time action, long acting, prolonged action, repeated action, slowing acting, sustained action, and sustained-action medications. Further discussions of these terms can be found in Lesczek Krowczynski, Extended-Release Dosage Forms, 1987 (CRC Press, Inc.).
  • Controlled release technologies cover a very broad spectrum of drug dosage forms. Controlled release technologies include, but are not limited to physical systems and chemical systems.
  • Physical systems include, but are not limited to, reservoir systems with rate-controlling membranes, such as microencapsulation, macroencapsulation, and membrane systems; reservoir systems without rate-controlling membranes, such as hollow fibers, ultra microporous cellulose triacetate, and porous polymeric substrates and foams; monolithic systems, including those systems physically dissolved in non-porous, polymeric, or elastomeric matrices (e.g., nonerodible, erodible, environmental agent ingression, and degradable), and materials physically dispersed in non-porous, polymeric, or elastomeric matrices (e.g., nonerodible, erodible, environmental agent ingression, and degradable); laminated structures, including reservoir layers chemically similar or dissimilar to outer control layers; and other physical methods, such as osmotic pumps, or adsorption onto ion-exchange resins.
  • rate-controlling membranes such as microencapsulation, macroencapsulation, and membrane systems
  • Chemical systems include, but are not limited to, chemical erosion of polymer matrices (e.g., heterogeneous, or homogeneous erosion), or biological erosion of a polymer matrix (e.g., heterogeneous, or homogeneous). Additional discussion of categories of systems for controlled release can be found in Agis F. Kydonieus, Controlled Release Technologies: Methods, Theory and Applications, 1980 (CRC Press, Inc.).
  • controlled release drug formulations that are developed for oral administration. These include, but are not limited to, osmotic pressure-controlled gastrointestinal delivery systems; hydrodynamic pressure-controlled gastrointestinal delivery systems; membrane permeation-controlled gastrointestinal delivery systems, which include microporous membrane permeation-controlled gastrointestinal delivery devices; gastric fluid-resistant intestine targeted controlled-release gastrointestinal delivery devices; gel diffusion-controlled gastrointestinal delivery systems; and ion-exchange-controlled gastrointestinal delivery systems, which include cationic and anionic drugs. Additional information regarding controlled release drug delivery systems can be found in Yie W. Chien, Novel Drug Delivery Systems, 1992 (Marcel Dekker, Inc.).
  • a suitable dosage can be determined by an attending physician or other qualified medical personnel, based on various clinical factors. As is well known in the medical arts, dosages for any one patient depend upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex of the patient, time, and route of administration, general health, and other drugs being administered concurrently.
  • a subject antibody can be administered in amounts from 1 ng/kg body weight to 20 mg/kg body weight per dose, for example from 0.001 to 10, 0.01 to 10, 0.1 to 10, 1 to 10, 0.001 to 1, 0.01 to 1, 0.1 to 1, 0.05 to 5, 0.05 to 0.5, or 0.5 to 5 mg/kg body weight. However, doses below or above this exemplary range are envisioned, especially considering the aforementioned factors. If the regimen is a continuous infusion, it may be in the range of 1 pg to 10 mg/kg of body weight per minute.
  • a dose of a subject anti-LAG-3 antibody is in the range of 0.001 ⁇ g to 100 mg, for example 0.001 ⁇ g to 10 mg, 0.001 ⁇ g to 1 mg, 0.001 ⁇ g to 0.1 mg, 0.01 ⁇ g to 10 mg, 0.1 ⁇ g to 10 mg, 0.1 ⁇ g to 1 mg, or 0.1 ⁇ g to 0.1 mg, or 0.01 to 100 mg, 0.01 to 10 mg, 0.01 to 1 mg, 0.01 to 0.1 mg, 0.1 to 100 mg, 0.1 to 10 mg, 0.1 to 1 mg.
  • the dosage can range, for example from about 0.0001 to 100 mg/kg, or from about 0.01 to 5 mg/kg (e.g., 0.02 to 5 mg/kg, 0.25 to 5 mg/kg, 0.5 to 5 mg/kg, 0.75 to 5 mg/kg, 1 to 5 mg/kg, 2 to 5 mg/kg, etc.) body weight.
  • dosages can be 0.1, 1, or 10 mg/kg body weight or within the range of 0.01-10 mg/kg, or at least 0.1 mg/kg.
  • a dose of an anti-LAG-3 antibody of the invention, or fragment thereof is up to 0.5 mg/kg body weight, for example in the range of 0.0001 to 0.5 mg/kg, 0.001 to 0.5 mg/kg, 0.01 to 0.5 mg/kg, 0.1 to 0.5 mg/kg, 0.0001 to 0.1 mg/kg, 0.001 to 0.1 mg/kg, 0.01 to 0.1 mg/kg body weight.
  • a subject anti-LAG-3 antibody is administered in an amount that provides for a peak serum concentration of from about 0.001 ⁇ g/ml to about 1 mg/ml, for example 0.0001 ⁇ g/ml to 1 ⁇ g/ml, 0.001 ⁇ g/ml to 1 ⁇ g/ml, 0.001 ⁇ g/ml to 0.1 ⁇ g/ml, 0.01 to 1, or 0.01 to 0.1 ⁇ g/ml, or from about 0.005 ⁇ g/ml to about 1 ⁇ g/ml, or from about 0.1 ⁇ g/ml to about 1 ⁇ g/ml, or from about 1 ⁇ g/ml to about 2.5 ⁇ g/ml, from about 2.5 ⁇ g/ml to about 5 pg/ml, from about 5 ⁇ g/ml to about 7.5 ⁇ g/ml, from about 7.5 ⁇ g/ml to about 10 ⁇ g/ml, from about 10 ⁇ g/ml to about 25
  • a subject anti-LAG-3 antibody is administered in an amount that provides for a peak serum concentration of greater than 1 mg/ml, e.g., from about 1 mg/ml to about 2 mg/ml, from about 2 mg/ml to about 5 mg/ml, or from about 5 mg/ml to about 10 mg/ml.
  • an anti-LAG-3 antibody of the invention, or fragment thereof is administered in an amount that provides for a peak serum concentration of up to 1 ⁇ g/ml, for example in the range of 0.0001 ⁇ g/ml to 1 ⁇ g/ml, 0.001 pg/ml to 1 ⁇ g/ml, 0.01 ⁇ g/ml to 1 ⁇ g/ml, 0.1 to 1 ⁇ g/ml, 0.0001 ⁇ g/ml to 0.1 ⁇ g/ml, 0.001 pg/ml to 0.1 ⁇ g/ml, or 0.01 ⁇ g/ml to 0.1 ⁇ g/ml.
  • such administration is by subcutaneous injection.
  • An exemplary treatment entails administration in multiple dosages over a prolonged period, for example, of at least six months. Additional exemplary treatment regimens entail administration once per every two weeks or once a month or once every 3 to 6 months.
  • Exemplary dosage schedules include 0.01 to 1 mg/kg, 0.01 to 0.1 mg/kg, 0.1 to 1 mg/kg, 1 to 10 mg/kg or 15 mg/kg on consecutive days, 0.02 to 20 mg/kg, for example 0.2 mg/kg, 0.5 mg/kg, 1 mg/kg, 5 mg/kg, 10 mg/kg, or 20 mg/kg on alternate days, or 0.1 to 100 mg/kg, for example 1 mg/kg, 10 mg/kg, 20 mg/kg, 30 mg/kg, 40 mg/kg, 50 mg/kg, or 60 mg/kg weekly.
  • two or more monoclonal antibodies with different binding specificities are administered simultaneously, in which case the dosage of each antibody administered falls within the ranges indicated. Progress can be monitored by periodic assessment.
  • the number of CD4+ and/or CD8+ T cells expressing LAG-3 in a subject is relatively low. It is expected that a single administration of an antibody of the invention (in particular, an antibody of the invention that has a serum half-life of at least two weeks and that lacks significant CDC and ADCC activity, such as a human IgG isotype antibody (which lacks CDC and ADCC activity), or an antibody that comprises one or more mutations to reduce or abolish CDC and ADCC activity) may be effective at inhibiting antigen-induced CD4+ and/or CD8+ T cell proliferation for at least several weeks.
  • an antibody of the invention in particular, an antibody of the invention that has a serum half-life of at least two weeks and that lacks significant CDC and ADCC activity, such as a human IgG isotype antibody (which lacks CDC and ADCC activity), or an antibody that comprises one or more mutations to reduce or abolish CDC and ADCC activity
  • a suitable treatment regimen may comprise administration of an antibody of the invention (for example 0.01 to 1 mg/kg of the antibody) once every four, six, eight, or ten weeks, or once every two or three months.
  • Such treatment may be provided over a period of at least six months, or at least one, two, three, four, or five years, or longer, for example throughout the course of a disease that is treated by the administration, or throughout the lifetime of the subject.
  • dose levels and administration schedules can vary as a function of the specific antibody, the severity of the symptoms and the susceptibility of the subject to side effects.
  • Preferred dosages and administration schedules for a given compound are readily determinable by those of skill in the art by a variety of means.
  • a subject antibody is administered to an individual using any available method and route suitable for drug delivery, including in vivo and ex vivo methods, as well as systemic and localized routes of administration.
  • routes of administration include intranasal, intramuscular, intratracheal, intrathecal, intracranial, subcutaneous, intradermal, topical, intravenous, intraperitoneal, intraarterial (e.g., via the carotid artery), spinal or brain delivery, rectal, nasal, oral, and other enteral and parenteral routes of administration. Routes of administration can be combined, if desired, or adjusted depending upon the antibody and/or the desired effect.
  • a subject antibody composition can be administered in a single dose or in multiple doses. In some embodiments, a subject antibody composition is administered orally. In some embodiments, a subject antibody composition is administered via an inhalational route. In some embodiments, a subject antibody composition is administered intranasally.
  • a subject antibody composition is administered locally. In some embodiments, a subject antibody composition is administered intracranially. In some embodiments, a subject antibody composition is administered intravenously. In some embodiments, a subject antibody composition is administered intrathecally.
  • An antibody of the present disclosure can be administered to a host using any available conventional methods and routes suitable for delivery of conventional drugs, including systemic or localized routes.
  • routes of administration contemplated by the invention include, but are not necessarily limited to, enteral, parenteral, or inhalational routes.
  • Parenteral routes of administration other than inhalation administration include, but are not necessarily limited to, topical, transdermal, subcutaneous, intramuscular, intraorbital, intracapsular, intraspinal, intrasternal, intrathecal, and intravenous routes, i.e., any route of administration other than through the alimentary canal.
  • Parenteral administration can be carried to effect systemic or local delivery of a subject antibody. Where systemic delivery is desired, administration typically involves invasive or systemically absorbed topical or mucosal administration of pharmaceutical preparations.
  • a subject antibody can also be delivered to the subject by enteral administration.
  • Enteral routes of administration include, but are not necessarily limited to, oral and rectal (e.g., using a suppository) delivery.
  • treatment is meant at least an amelioration of the symptoms associated with the pathological condition afflicting the host, where amelioration is used in a broad sense to refer to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the pathological condition being treated, such as an immune disorder.
  • amelioration also includes situations where the pathological condition, or at least symptoms associated therewith, are completely inhibited, e.g. prevented from happening, or stopped, e.g. terminated, such that the host no longer suffers from the pathological condition, or at least the symptoms that characterize the pathological condition.
  • a subject antibody is administered by injection and/or delivery, e.g., to a site in a brain artery or directly into brain tissue.
  • a subject antibody can also be administered directly to a target site e.g., by biolistic delivery to the target site.
  • hosts are treatable according to the subject methods.
  • hosts are “mammals” or “mammalian,” where these terms are used broadly to describe organisms which are within the class mammalia, including the orders carnivore (e.g., cats), herbivores (e.g., cattle, horses, and sheep), omnivores (e.g., dogs, goats, and pigs), rodentia (e.g., mice, guinea pigs, and rats), and primates (e.g., humans, chimpanzees, and monkeys).
  • carnivore e.g., cats
  • herbivores e.g., cattle, horses, and sheep
  • omnivores e.g., dogs, goats, and pigs
  • rodentia e.g., mice, guinea pigs, and rats
  • primates e.g., humans, chimpanzees, and monkeys.
  • the host is an individual that has a complement system, such as a mammal, fish, or invertebrate.
  • a complement system such as a mammal, fish, or invertebrate.
  • the host is a complement system-containing mammal, fish, or invertebrate companion animal, agricultural animal, work animal, zoo animal, or lab animal.
  • the host is human.
  • compositions comprising a container suitable for containing a composition comprising a subject anti-LAG-3 antibody for administration to an individual.
  • a subject antibody can be disposed within a container suitable for containing a pharmaceutical composition.
  • the container can be, for example, a bottle (e.g., with a closure device, such as a cap), a blister pack (e.g., which can provide for enclosure of one or more doses per blister), a vial, flexible packaging (e.g., sealed Mylar or plastic bags), an ampule (for single doses in solution), a dropper, a syringe, thin film, a tube and the like.
  • a container such as a sterile container, comprises a subject pharmaceutical composition.
  • the container is a bottle or a syringe.
  • the container is a bottle.
  • the container is a syringe.
  • Kits with unit doses of a subject antibody e.g. in oral or injectable doses, are provided.
  • kits in addition to the containers containing the unit doses will be an informational package insert describing the use and attendant benefits of the antibody in treating pathological condition of interest.
  • Preferred compounds and unit doses are those described herein above.
  • an antibody of the invention or antigen-binding fragment thereof, or a pharmaceutical composition of the invention, for use as a medicament.
  • An antibody, fragment, or composition of the present invention may be utilised in any therapy where it is desired to increase the effects of LAG-3 in the subject.
  • subject includes any human or nonhuman animal.
  • nonhuman animal includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, cows, horses, chickens, amphibians, and reptiles, although mammals are preferred, such as non-human primates, sheep, dogs, cats, cows and horses.
  • the antibody, fragment, or composition may be used in any therapy where it is desired to negatively regulate T cell proliferation and/or function.
  • an antibody of the invention, or antigen-binding fragment thereof, or a pharmaceutical composition of the invention for use in the treatment of a disorder associated with proliferation or activity of CD4+ and/or CD8+ T cells in a subject, or a disorder associated with decreased expression and/or activity of LAG-3 in a subject.
  • an antibody of the invention or antigen-binding fragment thereof, or a pharmaceutical composition of the invention, in the manufacture of a medicament for the treatment of a disorder associated with proliferation or activity of CD4+ and/or CD8+ T cells in a subject, or disorder associated with decreased expression and/or activity of LAG-3 in a subject.
  • a method of treating a disorder associated with proliferation or activity of CD4+ and/or CD8+ T cells in a subject, or disorder associated with decreased expression and/or activity of LAG-3 in a subject which comprises administering an effective amount of an antibody of the invention, or antigen-binding fragment thereof, or a pharmaceutical composition of the invention, to a subject in need of such treatment.
  • the disorder associated with proliferation or activity of CD4+ and/or CD8+ T cells may be an immune disorder, in particular a T-cell-mediated immune disorder, such as an inflammatory disease, or an autoimmune disorder.
  • the antibody, fragment or composition may be used to reduce the inflammatory process or to prevent the inflammatory process.
  • an in vivo reduction of T cell proliferation or activation in particular those involved in inappropriate inflammatory immune responses, for example recruited to the vicinity/location of such a response.
  • Reduction of T cell proliferation or activation may be a reduction of 10, 20, 30, 40, 50, 60, 70, 80, 90 or more percent in comparison to before treatment or without treatment.
  • treatment with an antibody, fragment or composition according to the present invention may allow a reduction in the T cell proliferation or activation, without reducing the patient's general level of T cells (unactivated T cells). This may result in fewer side effects, and prevent T cell depletion in the patient.
  • the immune disorder may, for example be selected from the group consisting of infections (viral, bacterial, fungal and parasitic), endotoxic shock associated with infection, sepsis, arthritis, rheumatoid arthritis, asthma, COPD, pelvic inflammatory disease, Alzheimer's Disease, inflammatory bowel disease, Crohn's disease, ulcerative colitis, Peyronie's Disease, coeliac disease, gallbladder disease, Pilonidal disease, peritonitis, psoriasis, vasculitis, surgical adhesions, stroke, Type I Diabetes, lyme disease, arthritis, meningoencephalitis, autoimmune uveitis, immune mediated inflammatory disorders of the central and peripheral nervous system such as multiple sclerosis, lupus (such as systemic lupus erythematosus) and Guillain-Barré syndrome, Atopic dermatitis, autoimmune hepatitis, fibrosing alveolitis, Grave's disease, IgA nephro
  • An antibody, fragment, or composition of the invention may be used in any therapy where it is desired to inhibit binding of LAG-3 to MHC class II molecules, to antagonise MHC class II-activating signal into antigen-presenting cells (APCs), or to inhibit LAG-3-induced APC activation.
  • APCs antigen-presenting cells
  • an antibody of the invention or antigen-binding fragment thereof, or a pharmaceutical composition of the invention, for use in the treatment of a disorder associated with activation of APCs in a subject.
  • an antibody of the invention or antigen-binding fragment thereof, or a pharmaceutical composition of the invention, in the manufacture of a medicament for the treatment of a disorder associated with activation of APCs in a subject.
  • a method of treating a disorder associated with activation of APCs in a subject which comprises administering an effective amount of an antibody of the invention, or antigen-binding fragment thereof, or a pharmaceutical composition of the invention, to a subject in need of such treatment.
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect can be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or can be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease.
  • Treatment covers any treatment of a disease in a mammal, particularly in a human, and includes: (a) preventing the disease from occurring in a subject which can be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., causing regression of the disease.
  • nonhuman animal includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, cows, horses, chickens, amphibians, and reptiles, although mammals are preferred, such as non-human primates, sheep, dogs, cats, cows and horses.
  • a “therapeutically effective amount” or “efficacious amount” refers to the amount of an anti-LAG-3 antibody that, when administered to a mammal or other subject for treating a disease, is sufficient to effect such treatment for the disease.
  • the “therapeutically effective amount” will vary depending on the anti-LAG-3 antibody, the disease and its severity and the age, weight, etc., of the subject to be treated.
  • FIG. 1 shows amino acid sequence (SEQ ID NO:27) of mature human LAG-3 protein.
  • the four extracellular Ig superfamily domains are at amino acid residues: 1-149 (D1); 150-239 (D2); 240-330 (D3); and 331-412 (D4).
  • the amino acid sequence of the extra-loop structure of the D1 domain of human LAG-3 protein is shown underlined in bold (SEQ ID NO:40);
  • FIG. 2 shows a graphical representation of the V H CDR loops of monoclonal antibody 13E2 (Lefranc, M.-P. et al., Dev. Comp. Immunol., 27, 55-77 (2003));
  • FIG. 3 shows the amino acid sequence of the V H domain of monoclonal antibody 13E2 aligned with an encoding nucleic acid sequence
  • FIG. 4 shows a graphical representation of the V L CDR loops of monoclonal antibody 13E2 (Lefranc, M.-P. et al., Dev. Comp. Immunol., 27, 55-77 (2003));
  • FIG. 5 shows the amino acid sequence of the V L domain of monoclonal antibody 13E2 aligned with an encoding nucleic acid sequence
  • FIG. 6 shows a graphical representation of the V H CDR loops of monoclonal antibody 34F4 (Lefranc, M.-P. et al., Dev. Comp. Immunol., 27, 55-77 (2003));
  • FIG. 7 shows the amino acid sequence of the V H domain of monoclonal antibody 34F4 aligned with an encoding nucleic acid sequence
  • FIG. 8 shows a graphical representation of the V L CDR loops of monoclonal antibody 34F4 (Lefranc, M.-P. et al., Dev. Comp. Immunol., 27, 55-77 (2003));
  • FIG. 9 shows the amino acid sequence of the V L domain of monoclonal antibody 34F4 aligned with an encoding nucleic acid sequence
  • FIG. 10 shows the top V-D-J germline BLAST alignments for nucleotide sequence encoding the V H region of monoclonal antibody 13E2;
  • FIG. 11 shows the top V-J germline BLAST alignments for nucleotide sequence encoding the V L region of monoclonal antibody 13E2;
  • FIG. 12 shows the top V-D-J germline BLAST alignments for nucleotide sequence encoding the V H region of monoclonal antibody 34F4;
  • FIG. 13 shows the top V-J germline BLAST alignments for nucleotide sequence encoding the V L region of monoclonal antibody 34F4;
  • FIG. 14 shows the results of binding of different concentrations of agonistic anti-LAG-3 monoclonal antibodies 13E2 and 34F4, and antagonistic anti-LAG-3 monoclonal antibody 1764, to LAG-3-transfected Chinese hamster ovary (CHO) cells, compared to an isotype control antibody (mIgG1);
  • FIG. 15 shows the results of binding of different concentrations of agonistic anti-LAG-3 monoclonal antibodies 13E2 and 34F4, and antagonistic anti-LAG-3 monoclonal antibody 1764, to CD4 + and CD8 + primary cells (SEB-stimulated PBMCs), compared to an isotype control antibody (mIgG1), from a healthy donor (donor 1);
  • FIG. 16 A shows the results of inhibition of binding of IMP321 (LAG-3Ig, 1 ⁇ g/ml) to MHC class II-positive B cells by different concentrations of agonistic anti-LAG-3 monoclonal antibodies 13E2 and 34F4, and antagonistic anti-LAG-3 monoclonal antibody 1764, compared to an isotype control antibody (mIgG1).
  • FIG. 16 B shows the results of inhibition of activation of THP-1 cells by IMP321 (20 ng/ml) in the presence of different concentrations of agonistic anti-LAG-3 monoclonal antibodies 13E2 and 34F4, and antagonistic anti-LAG-3 monoclonal antibody 1764, compared to an isotype control antibody (mIgG1);
  • FIG. 17 (A) illustrates the CMV-induced proliferation profiles of the CD8 + T cells of one donor in the presence of mIgG1, 17B4, 13E2 or 34F4, analysed by flow cytometry and the gating strategy used for the assay described in Example 10.
  • FIG. 17 (B) shows the results of an assay for the inhibition of CD8 + T cell proliferation by antibodies 13E2, 34F4, and 1764, compared with the isotype control antibody (mIgG1) for the same donor. The baseline proliferation is also shown (No Stim);
  • FIG. 18 shows the results of an assay for the inhibition of CD4 + or CD8 + T cell proliferation by antibodies 13E2 or 34F4, compared with the isotype control antibody (mIgG1), in several different donors;
  • FIG. 19 shows the effect of different concentrations of agonistic anti-LAG-3 monoclonal antibodies 13E2 and 34F4 on CD8 + T cell proliferation
  • FIG. 20 shows the heavy chain amino acid sequence of a chimeric 13E2-human IgG4 Fc antibody (the chimeric 13E2-human IgG4 Fc heavy chain amino acid sequence is referred to as 13E2IgG4mut in the figure), and the light chain amino acid sequence of a chimeric 13E2-human IgK antibody (the chimeric 13E2-human IgK light chain amino acid sequence is referred to as 13E2IgK in the figure);
  • FIG. 21 illustrates the different effects on T cells of depleting, antagonist, and agonist anti-LAG-3 antibodies
  • FIG. 22 shows an alignment of the variable regions of humanized VH variants 1-4 (VH 1 , VH 2 , VH 3 , and VH 4 ), and an alignment of the variable regions of humanized VL variants 1-4 (VL 1 , VL 2 , VL 3 , and VL 4 ), with the corresponding sequence of the original mouse monoclonal antibody 13E2 (13E2 VH and 13E2 VL, respectively).
  • CDR sequences are highlighted in grey. Changes in the humanized framework sequences of the variants, compared with the original mouse sequence, are shown underlined and in bold;
  • FIG. 23 shows the heavy chain amino acid sequence of a humanized 13E2-human IgG4 Fc antibody (IMP761) aligned with the chimeric 13E2-human IgG4 Fc heavy chain amino acid sequence (13E2IgG4mut) of antibody Chim13E2IgG4.
  • the V H region is shown in bold, and the Fc region is shown highlighted.
  • Amino acid residues of the humanized IMP761 sequence that differ from corresponding residues of the chimeric 13E2IgG4mut sequence are single underlined.
  • CDR sequences (based on combined IMGT/Kabat CDR sequence identification) are shown double underlined;
  • FIG. 24 shows the light chain amino acid sequence of a humanized 13E2-human IgK antibody (IMP761) aligned with the chimeric 13E2-human IgK light chain amino acid sequence (13E2IgK) of antibody Chim13E2IgG4.
  • the V L region is shown in bold, and the IgK region is shown highlighted.
  • Amino acid residues of the humanized IMP761 sequence that differ from corresponding residues of the chimeric 13E2IgK sequence are single underlined.
  • CDR sequences (based on the combined IMGT/Kabat CDR sequence identification) are shown double underlined;
  • FIG. 25 shows the results of an assay to test binding of the chimeric 13E2-human IgG4 Fc antibody (Chim13E2IgG4) and IMP761 to CHO-LAG-3+ cells;
  • FIG. 26 shows the results of an assay to test the effect of IMP761 and Chim13E2IgG4 on: (a) antigen-induced CD8 + T-cell proliferation; and (b) CD25 expression within CD8 + T cells;
  • FIG. 27 shows the effect of IMP761 and Chim13E2IgG4 on antigen-induced CD8 + T-cell proliferation, and CD25 expression, as a plot of: (a) the percentage inhibition of CD8 + T-cell proliferation; and (b) the percentage inhibition of CD25 expression within CD8 + T cells, compared to an isotype-matched control;
  • FIG. 28 shows the effect of different concentrations of IMP761 and Chim13E2IgG4 on antigen-induced CD8 + T-cell proliferation
  • FIG. 29 shows the results of ADCC assays to determine whether IMP761 has cytotoxic activity against LAG-3-expressing cells, using: an ADCC Reporter Bioassay available from Promega (a); and an ADCC assay using IL-2-stimulated PBMCs as effector cells—the results are plotted as the percentage of: CD4 + and CD8 + T cells (b); or LAG-3+CD4 + and LAG-3+CD8 + T cells (c) in the PBMC population;
  • FIG. 30 shows the results of CDC assays using rabbit complement to determine whether IMP761 has cytotoxic activity against LAG-3-expressing cells—the results are plotted as the percentage of: (a) CD4 + and CD8 + T cells; or (b) LAG-3+CD4 + and LAG-3+CD8 + T cells, in the PBMC population; and
  • FIG. 31 shows the results of an assay to determine whether IMP761 has cytotoxic activity against LAG-3-expressing cells after culturing antigen-stimulated PBMCs with the antibody for 3 days.
  • the results are plotted as: (a) the percentage of CD4 + and CD8 + T cells in the PBMC population; and (b) the percentage of LAG-3+ cells in the CD4 + and CD8 + T cell subpopulation.
  • mice numbers 1-15 below were immunised according to the immunisation protocol described below.
  • mice Thirteen mice were each immunised with subcutaneous (s.c.) injections of 100 ⁇ g of IMP321 (LAG-3Ig), clinical grade lot S017/LC1/041011 (termed “LC1” below): 3 times (mouse no. 12); 4 times (mouse no. 9); 5 times (mice nos. 5 and 14); or 6 times (mice nos. 3, and 11) at Day 0, Day 15, Day 30, Day 50, Day 67 and Day 108. Mice nos. 1, 2, 4, 8, 10, 13 and 15 were immunised up to 4 additional times. In parallel, two mice (mouse no. 6 and mouse no.
  • CFA Complete Freund's Adjuvant
  • IFA Incomplete Freund's Adjuvant
  • mice nos. 1, 2, 3, 4, 8, 10, 11, 13 and 15 were drawn (mice nos. 5, 9, 12 and 14 had already been sacrificed) and analyzed in an Enzyme-Linked Immunosorbent Assay (ELISA) using LAG-3 D1-D4 as coated antigen, purified anti-LAG-3 17B4 murine monoclonal antibody as reference, and a goat anti-mouse Ig-HRP as labelled secondary antibody, to determine the concentration of anti-LAG-3 antibodies present in the serum.
  • ELISA Enzyme-Linked Immunosorbent Assay
  • the same serum samples were assessed for their ability to inhibit the binding of IMP321 to its ligand, MHC class II, expressed on Raji B cells.
  • Ten microliters of a solution of 10 ⁇ g/ml of Alexa-Fluor488-conjugated IMP321 was preincubated with and without 5 ⁇ l of the serum collected from each mouse or na ⁇ ve serum for 30 min at 4° C.
  • Raji cells were added to a final volume of 50 ⁇ l and incubated for 30 minutes at 4° C. The cell-bound fluorescence was analyzed by flow cytometry.
  • Mouse no. 3 was selected because the serum from this mouse showed a high titre (423 ⁇ g/ml) in the D1-D4 serum ELISA assay (Table 1), and a strong capacity to inhibit the binding of IMP321 to MHC class II + Raji B cells.
  • mouse no. 3 received an intravenous (i.v.) boost with 10 ⁇ g D1-D4 LAG-3 (with no Fc tail) recombinant protein (produced in CHO cells and purified).
  • i.v. boost injection mouse no. 3 was sacrificed and the spleen was removed. The splenocytes were extracted by squeezing the pieces of spleen with a 5 ml syringe rubber plunger in a Petri dish containing complete serum-free DMEM.
  • the splenocytes and Sp2/0 myeloma cells were washed in serum-free medium.
  • the two cell types were mixed together in a ratio of 5:1 of splenocytes:myeloma, and then pelleted by centrifugation.
  • the fusion agent PEG-1500, Polyethylene Glycol solution 50% w/v in PBS, Roche 10783641001, 1 ml per 10 8 cells
  • the fusion agent PEG-1500, Polyethylene Glycol solution 50% w/v in PBS, Roche 10783641001, 1 ml per 10 8 cells
  • the cells were very gently resuspended, and diluted by doubling the volume after 90 seconds.
  • the cells were further diluted at regular intervals to a final dilution of 1 to 15 over approximately 5 minutes.
  • the cells were then centrifuged and resuspended in medium containing 10% FBS and incubated for approximately one hour at 37° C.
  • the cells (10,000 cells/well) were then plated in 46 96-well plates in complete RPMI containing 10% of Ultralow Ig FBS (Gibco 16250), 2% HAT (Gibco 21060) and supplemented with 10% BM Condimed H1 (a supplement to the culture medium to support the growth of B-cell hybridomas after fusion and during cloning, Roche 11088947001), and cultured until screening.
  • the screening was performed by cytometry using CHO cells, with membrane-expressed LAG-3, to analyse the binding ability of antibodies present in the supernatants of growing hydridomas, revealed by a FITC-conjugated goat-anti mouse Ig.
  • the positive hybridomas were expanded and rescreened on CHO LAG-3 + cells, and wild type CHO cells. From this fusion, a total of 632 wells were screened by FACS analysis on LAG-3 expressing CHO cells (a yield of 14%). 4 hybridoma clones were found to stably express anti-LAG-3 antibody, including 13E2. The hybridoma was then subcloned by limiting dilution.
  • SEQ ID NO:7 is the amino acid sequence of the heavy chain variable (V H ) domain of antibody 13E2.
  • the Complementarity Determining Regions (CDRs), as determined by the IMGT numbering system (Lefranc, M.-P. et al., Nucleic Acids Research, 27, 209-212 (1999)), are underlined.
  • the CDRs as determined by the Kabat numbering system, are shown in bold.
  • FIG. 2 shows a graphical representation of the V H CDR loops of monoclonal antibody 13E2 (Lefranc, M.-P. et al., Dev. Comp. Immunol., 27, 55-77 (2003)).
  • Shaded circles (residue nos. 4, 12, 13, 19, 21, 23, 25, 41, 50, 52, 53, 71, 76, 78, 87, 89, 91, 94, 100) are hydrophobic (non-polar) residues in frameworks 1-3 at sites that are hydrophobic in the majority of antibodies. Squares are key residues at the start and end of each CDR.
  • Amino acid residue nos. 23, 41, 89, 104, 118 in the framework are structurally conserved amino acids;
  • SEQ ID NO:8 is the amino acid sequence of the light chain variable (V L ) domain of antibody 13E2.
  • the Complementarity Determining Regions (CDRs), as determined by the IMGT numbering system (Lefranc, M.-P. et al., Nucleic Acids Research, 27, 209-212 (1999)), are underlined.
  • the CDRs as determined by the Kabat numbering system, are shown in bold.
  • FIG. 4 shows a graphical representation of the V L CDR loops of monoclonal antibody 13E2 (Lefranc, M.-P. et al., Dev. Comp. Immunol., 27, 55-77 (2003)).
  • Shaded circles (residue nos. 4, 11, 19, 21, 23, 25, 40, 41, 52, 53, 54, 71, 76, 87, 89, 91, 94, 96, 100, 101) are hydrophobic (non-polar) residues in frameworks 1-3 at sites that are hydrophobic in the majority of antibodies. Squares are key residues at the start and end of each CDR.
  • Amino acids residue nos. 23, 41, 89, 104, 118 in the framework are structurally conserved amino acids.
  • Nucleic acid sequence encoding the VH domain of monoclonal antibody 13E2 is shown in FIG. 3 , and below:
  • nucleic acid BLAST alignment shows that nucleic acid sequence encoding the VH domain of monoclonal antibody 13E2 has significant identity to the sequence of the following germline genes: IGHV8-8*01, IGHV8-11*01, IGHV8-12*01, IGHD2-12*01, IGHD1-1*01, IGHJ1*01, IGHJ1*02, IGHJ1*03.
  • FIG. 10 shows an alignment of nucleic acid sequence encoding the VH domain of the 13E2 antibody with its top germline gene match.
  • FIG. 10 shows that a portion comprising nucleotides 1-301 of the 13E2 VH region (which encompasses heavy chain framework regions FR1, FR2, and FR3) has 92.7% nucleic acid sequence identity with nucleic acid sequence of V gene IGHV8-8*01.
  • Nucleic acid sequence encoding the VL domain of monoclonal antibody 13E2 is shown in FIG. 5 , and below:
  • nucleic acid BLAST alignment shows that nucleic acid sequence encoding the VL domain of monoclonal antibody 13E2 has significant identity to the sequence of the following germline genes: IGKV6-17*01, IGKV6-25*01, IGKV6-23*01, IGKJ2*01, IGKJ2*03, IGKJ2*02.
  • FIG. 11 shows an alignment of nucleic acid sequence encoding the VL domain of the 13E2 antibody with its top germline gene match.
  • FIG. 11 shows that a portion comprising nucleotides 1-284 of the 13E2 VL region (which encompasses light chain framework regions FR1, FR2, and FR3) has 94.7% nucleic acid sequence identity with nucleic acid sequence of V gene IGKV6-17*01.
  • mice nos. 1-4 Balb/c mice (referred to as mice nos. 1-4 below) were each immunised with four (mice nos. 2 and 4), or five (mice nos. 1 and 3) s.c. injections of 100 ⁇ g IMP321 (LAG-3Ig), clinical grade lot S017/LC1/041011 (termed “LC1” below), at Day 0, Day 14, Day 28, Day 43 and Day 70.
  • An additional Balb/c mouse (referred to as mouse no. 5 below) was immunized with three s.c. injections of D1-D4 LAG-3.
  • the ability of the serum from each mouse to inhibit binding of IMP321 to the Raji B cells was determined by FACS analysis (as described in Example 1).
  • splenocytes from mouse no. 3 were fused with 15 million Sp2/0 myeloma cells following the same procedure described in Example 1.
  • the wells of 40 96-well plates were seeded with approximately 25,500 cells per well, and then cultured with supplementation of the culture medium with 10% BM Condimed Ht 2256 wells were screened by FACS analysis on LAG-3 expressing CHO cells (a yield of 59%).
  • Two stable anti-LAG-3 hybridomas were selected, including 34F4 (see Table 8 below).
  • SEQ ID NO: 17 is the amino acid sequence of the heavy chain variable (V H ) domain of antibody 34F4.
  • the Complementarity Determining Regions (CDRs), as determined by the IMGT numbering system (Lefranc, M.-P. et al., Nucleic Acids Research, 27, 209-212 (1999)), are underlined. The CDRs, as determined by the Kabat numbering system, are shown in bold.
  • FIG. 6 shows a graphical representation of the V H CDR loops of monoclonal antibody 34F4 (Lefranc, M.-P. et al., Dev. Comp. Immunol., 27, 55-77 (2003)).
  • Shaded circles (residue nos. 4, 12, 13, 19, 21, 23, 25, 41, 50, 52, 53, 71, 76, 78, 87, 89, 91, 94, 100) are hydrophobic (non-polar) residues in frameworks 1-3 at sites that are hydrophobic in the majority of antibodies. Squares are key residues at the start and end of each CDR.
  • Amino acid residue nos. 23, 41, 89, 104, 118 in the framework are structurally conserved amino acids.
  • SEQ ID NO: 18 is the amino acid sequence of the light chain variable (V L ) domain of antibody 34F4.
  • the Complementarity Determining Regions (CDRs), as determined by the IMGT numbering system (Lefranc, M.-P. et al., Nucleic Acids Research, 27, 209-212 (1999)), are underlined.
  • the CDRs as determined by the Kabat numbering system, are shown in bold.
  • FIG. 8 shows a graphical representation of the V L CDR loops of monoclonal antibody 34F4 (Lefranc, M.-P. et al., Dev. Comp. Immunol., 27, 55-77 (2003)).
  • Shaded circles (residue nos. 4, 11, 19, 21, 23, 25, 40, 41, 52, 53, 54, 71, 76, 87, 89, 91, 94, 96, 100, 101) are hydrophobic (non-polar) residues in frameworks 1-3 at sites that are hydrophobic in the majority of antibodies. Squares are key residues at the start and end of each CDR.
  • Amino acids residue nos. 23, 41, 89, 104, 118 in the framework are structurally conserved amino acids.
  • Nucleic acid sequence encoding the VH domain of monoclonal antibody 34F4 is shown in FIG. 7 , and below:
  • nucleic acid BLAST alignment shows that nucleic acid sequence encoding the VH domain of monoclonal antibody 34F4 has significant identity to the sequence of the following germline genes: IGHV8-8*01, IGHV8-12*01, IGHV8-11*01, IGHD1-1*01, IGHD1-2*01, IGHD2-3*01, IGHJ2*01, IGHJ2*02, IGHJ2*03.
  • FIG. 12 shows an alignment of nucleic acid sequence encoding the VH domain of the 34F4 antibody with its top germline gene match.
  • FIG. 12 shows that a portion comprising nucleotides 1-301 of the 34F4 VH region (which encompasses heavy chain framework regions FR1, FR2, and FR3) has 94.4% nucleic acid sequence identity with nucleic acid sequence of V gene IGHV8-8*01.
  • Nucleic acid sequence encoding the VL domain of monoclonal antibody 34F4 is shown in FIG. 9 , and below:
  • nucleic acid BLAST alignment shows that nucleic acid sequence encoding the VL domain of monoclonal antibody 34F4 has significant identity to the sequence of the following germline genes: IGKV6-17*01, IGKV6-25*01, IGKV6-23*01, IGKJ1*01, IGKJ1*02, IGKJ2*01.
  • FIG. 13 shows an alignment of nucleic acid sequence encoding the VL domain of the 34F4 antibody with its top germline gene match.
  • FIG. 13 shows that a portion comprising nucleotides 1-284 of the 34F4 VL region (which encompasses light chain framework regions FR1, FR2, and FR3) has 94.7% nucleic acid sequence identity with nucleic acid sequence of V gene IGKV6-17*01.
  • LAG-3 + -transfected CHO cells, or SEB-stimulated PBMCs from a healthy donor were incubated with anti-LAG-3 monoclonal antibody, or an isotype control (mIgG1) for 30 minutes in PBS, BSA 0.5%, Azide 0.1% at 4° C.
  • Cells were washed, and cell-bound antibody was revealed by a FITC-conjugated goat F(ab′)2-anti-mouse Ig (H+L) (Coulter). The secondary antibody was washed away, and the CHO cells were directly analyzed by flow cytometry.
  • the PBMCs were phenotyped using CD4-PE-Cy7 and CD8-APC-Cy7.
  • results are presented as the mean of fluorescence intensity (MFI) of CHO cells transfected with human LAG-3-encoding plasmid as a function of antibody concentration. The results are shown in Table 12 below, and in FIG. 14 .
  • the EC 50 value for binding of each antibody to LAG-3-expressing CHO cells is: 1764: 0.7 nM; 13E2: 0.3 nM; 34F4: 0.5 nM.
  • the mean EC 50 values from four independent experiments (data not shown) for binding of each antibody to LAG-3-expressing CHO cells is: 1764: 0.7 nM; 13E2: 0.4 nM; 34F4: 0.5 nM.
  • the mean, from the four independent experiments, of the 13E2 EC 50 value is 2.7 times the mean of the 1764 EC 50 value.
  • the mean, from the four independent experiments, of the 34F4 EC 50 value is 1.6 times the mean of the 1764 EC 50 value.
  • the results are presented as the mean of fluorescence intensity on the CD4+ or CD8+ cells from PBMCs of a donor (Donor 1) stimulated for three days with 0.5 ⁇ g/ml SEB as a function of antibody concentration.
  • the results for binding to CD4 + and CD8 + cells, for Donor 1, are shown in Table 13 below, and in FIG. 15 .
  • the EC 50 value for binding of each antibody to CD4 + cells is: 1764: 0.5 nM; 13E2: 0.1 nM; 34F4: 0.1 nM.
  • the mean EC 50 value from three donors (data not shown) for binding of each antibody to CD4 + cells is: 1764: 0.8 nM; 13E2: 0.2 nM; 34F4: 0.2 nM.
  • the mean of the 13E2 and 34F4 CD4 + EC 50 values from the three donors is 3.8 times the mean of the 1764 EC 50 value.
  • the EC50 value for binding of each antibody to CD8 + cells is: 1764: 0.7 nM; 13E2: 0.3 nM; 34F4: 0.2 nM.
  • the mean EC 50 value from three donors (data not shown) for binding of each antibody to CD8 + cells is: 1764: 1 nM; 13E2: 0.4 nM; 34F4: 0.5 nM.
  • the mean of the 13E2 and 34F4 CD8+EC 50 values from the three donors is 2.5 times the mean of the 1764 EC 50 value.
  • Binding of an IMP321 conjugate (LAG-3Ig-Alexa 488) to MHC class II-positive B cells (Raji cells) was determined following pre-incubation of the conjugate (1 pg/ml at 4° C.) with an anti-LAG-3 monoclonal antibody (13E2, 34F4, or 1764), or an isotype control (mIgG1). Analysis of cell-bound fluorescence was carried out using fluorescence-activated cell sorting (FACS).
  • MFI mean fluorescence intensity
  • IMP321 (20 ng/ml) was preincubated with anti-LAG-3 monoclonal antibody 13E2, 34F4, or 17B4, or an isotype control (mIgG1), for 5 minutes at 37° C., before incubation of the mixture with THP-1 cells for 4 hours at 37° C.
  • the amount of CCL4 secretion by the THP-1 cells was used to determine the level of monocyte activation.
  • the CCL4 concentration (expressed in pg/ml) as a function of Ab concentration is shown in Table 15 below, and in FIG. 16 B .
  • PBMCs from 3 healthy donors (0.2 ⁇ 10 6 cells/well, at 1 ⁇ 10 6 /ml in complete RPMI+10% FBS) were labelled with Carboxyfluorescein succinimidyl ester (CFSE) and incubated with a pool of peptides covering the sequence of CMV pp35 in the presence of monoclonal anti-LAG-3 antibody 13E2, 34F4, 1764, or an isotype control (mIgG1) (supra-optimal dose, 300 ng/ml for donor #1 and #2, 100 ng/ml for donor #3).
  • CFSE Carboxyfluorescein succinimidyl ester
  • the T cell response was investigated by measuring the CFSE-based proliferation of CD4 + or CD8 + T cells on day 5.
  • the FACS profiles of the CD8 + T cells of donor #1 in the presence of each different antibody, as well as the gating strategy, are shown in FIG. 17 (A) .
  • FIG. 17 (B) shows the percentage of CD8 + T cells under each division peak, as a function of cell division, for the same donor.
  • the results for the 3 donors are shown in Table 16 below.
  • the baseline proliferation without antigenic peptides (No stim) was also measured (see FIG. 17 (A) lowest panel and Table 16).
  • the CD4 + T cells of donor #1 did not display any CMV-specific proliferation, so the results for this population are not included.
  • the Proliferation Index (calculated as the sum of: the percentage of CD4 + or CD8 + T cells under each division peak, multiplied by the division number) is provided in Table 17. This index emphasises the percentages for cells which have experienced several rounds of division. Table 17 also records the percentage inhibition for each antibody compared with the isotype control (mIgG1) based on the PI values.
  • PBMCs from 12 healthy donors were labelled with CFSE and incubated with a pool of peptides covering the sequence of CMV pp35 in the presence of monoclonal anti-LAG-3 antibody 13E2, 34F4, or an isotype control (mIgG1).
  • the T cell response was investigated by measuring the CFSE-based proliferation of CD4 + or CD8 + T cells on day 5.
  • the percentage of CD4 + or CD8 + T cells under each division peak was calculated as a function of cell division, using the gating strategy illustrated in FIG. 17 (A) .
  • the baseline proliferation without antigenic peptides (No stim) was also measured.
  • the CD4 + T cells of donors #1, #5, and #12 did not display any CMV-specific proliferation, so the results for these samples are not included.
  • the Proliferation Index (PI) (calculated as the sum of: the percentage of CD4 + or CD8 + T cells under each division peak, multiplied by the division number) for each donor is provided in Table 18, and the results are plotted in FIG. 18 .
  • Table 18 also records the percentage inhibition for each antibody compared with the isotype control (mIgG1) based on the PI values.
  • CFSE-labelled PBMCs were stimulated by CMV peptide, as described above, in the presence of various concentrations of agonist anti-LAG-3 monoclonal antibody 13E2, 34F4, or an isotype control (mIgG1).
  • the T cell response was investigated by measuring CFSE-based proliferation of CD8 + T cells on day 5.
  • the Proliferation Index (calculated as the sum of the percentage of CD8 + T cells under each division peak, multiplied by the division number) is provided in Table 20.
  • Table 21 below records the CD8 + T cell Proliferation Index as a function of antibody concentration. The results in Table 21 are plotted in FIG. 19 .
  • results show that a dose as low as 30 ng/ml of monoclonal anti-LAG-3 antibody 13E2, or 34F4, causes maximal inhibition of CD8 + T cell proliferation.
  • results also show that the effects of the antibodies were very similar.
  • CFSE-labelled PBMCs from 2 donors were stimulated by CMV peptide, as described above, in the presence of various concentrations of 34F4 antibody.
  • the 1 ⁇ g/ml dose of 34F4 was also assessed after neutralisation with a 10-fold excess of IMP321.
  • the T cell response was investigated by measuring the CFSE-based proliferation of CD8 + T cells on day 5.
  • the percentage of inhibition of the proliferation of CD8 + T cells was calculated based on the percentage of dividing cells observed in the presence of 34F4 antibody, or 34F4 antibody and IMP321 (LAG-3Ig), compared to a control with or without IMP321.
  • CFSE-labelled PBMCs were stimulated by CMV peptides, as described above, in the presence of 13E2 or 34F4 antibody, with or without IL-2.
  • the T cell response was investigated by measuring the CFSE-based proliferation of CD8 + T cells on day 5.
  • the percentage of inhibition of the proliferation of CD8 + T cells was calculated based of the percentage of dividing cells observed in the presence of 13E2 or 34F4 antibody, with or without or IL-2, compared to an isotype control with or without IL-2.
  • PBMCs Peripheral blood mononuclear cells
  • T cells lymphocytes
  • B cells and NK cells
  • monocytes monocytes
  • dendritic cells lymphocytes
  • IFN- ⁇ is predominantly secreted by activated CD4 + and CD8 + memory and effector T cells and by NK cells upon activation. After re-stimulation with specific antigen in vitro, secretion of IFN- ⁇ is induced.
  • PBMCs from four healthy donors 0.2 ⁇ 10 6 cells/well, at 1 ⁇ 10 6 /ml in complete
  • PBMCs from four healthy donors (0.2 ⁇ 10 6 cells/well, at 1 ⁇ 10 6 /ml in complete RPMI+10% FBS) were labelled with CFSE, and incubated with a pool of peptides covering the sequence of CMV pp35 in the presence of monoclonal anti-LAG-3 antibody 13E2 or 34F4, or an isotype control (mIgG1).
  • the T cell response was investigated by measuring the expression of CD25, as an activation marker, on CD8 + T cells on day 5.
  • Nucleotide sequences encoding the murine variable region of the 13E2 heavy and light chains were fused to sequences encoding the constant region of the human IgG4 heavy and Kappa light chains, respectively. These synthetic chimeric sequences were subcloned into an expression vector, and expressed in CHO cells grown in suspension.
  • the heavy chain amino acid sequence of a chimeric 13E2-human IgG4 Fc antibody is shown below, and in FIG. 20 (A) .
  • the antibody comprises the V H domain of mouse monoclonal antibody 13E2, and a human IgG4 Fc portion with an S228P mutation (to abolish Fab arm exchange) (13E2IgG4mut).
  • the V H region is shown in bold, and the Fc region is shown highlighted.
  • the light chain amino acid sequence of a chimeric 13E2-human IgK antibody is shown below, and in FIG. 20 (B) .
  • the antibody comprises the V L domain of monoclonal antibody 13E2, and a wild-type human Ig kappa (IgK) chain C portion (13E2IgK).
  • IgK human Ig kappa chain C portion
  • the V L region is shown in bold, and the IgK region is shown highlighted.
  • 13E2IgK (SEQ ID NO: 37) MVSSAQFLGLLLLCFQGTRCDIVMTQPHKFMSTSVEDRVTITCKASQDVI FDVAWYQQKPGQSPKLLIYSASSRVSGVPDRFTGSGSGTDFTFTISSVQA EDLAVYYCQQHYSTPYTFGGGTTLEIKRTVAAPSVFIFPPSDEQLKSGTA SVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLT LSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
  • a chimeric 13E2-human antibody (referred to as Chim13E2IgG4) comprises the chimeric heavy and light chains: 13E2IgG4mut; and 13E2IgK.
  • IMGT/Kabat CDR sequence identification was used to graft CDRs from murine 13E2 to human frameworks in order to obtain a humanized version of 13E2.
  • synthetic chimeric sequences were subcloned into an expression vector, and expressed in CHO cells grown in suspension.
  • IMP761 Heavy and light chain amino acid sequences of the humanized 13E2 monoclonal antibody (referred to as IMP761) are shown below. Variable domains are shown in bold, CDR sequences are shown underlined.
  • Amino acid sequence of IMP761 heavy chain (SEQ ID NO: 84) MGWTLVFLFLLSVTAGVHS QITLKESGPTLVKPTQTLTLTCTFS GFSLST SGMGLG WIRQPPGKTLEWLT HIWWDDIKRYNPDLRS RLSITKDTSKNQVV LTMTNMDPLDTGTYYC ARIVEGSYSSSYFDV WGQGTLVTVSS ASTKGPSV FPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQ SSGLYSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPP CPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWY VDGVEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGL PSSIEKTISKAKGQPREPQVYTLPPSQEEMTKN
  • FIG. 23 Alignment of this sequence (IMP761 heavy chain) with the chimeric heavy chain sequence, 13E2IgG4mut, of Example 17 is shown in FIG. 23 .
  • the V H region is shown in bold, and the Fc region is shown highlighted.
  • Amino acid residues of the humanized IMP761 sequence that differ from corresponding residues of the chimeric 13E2IgG4mut sequence are single underlined.
  • CDR sequences (based on the combined IMGT/Kabat CDR sequence identification) are shown double underlined.
  • the changed residues in the humanized sequence are also set out in Table 26 below (as VH variant 4, VH 4 , as well as the changed residues in three other humanized variants of the original 13E2 heavy chain sequence: VH variants 1, 2, and 3, VH 1 , VH 2 , and VH 3 ).
  • the heavy chain framework sequences of the humanized antibody are:
  • VH FR1 QITLKESGPTLVKPTQTLTLTCTFS
  • VH FR2 WIRQPPGKTLEWLT
  • VH FR3 RLSITKDTSKNQVVLTMTNMDPLDTGTYYC
  • SEQ ID NO: 67 VH FR4: WGQGTLVTVSS.
  • Amino acid sequence of IMP761 light chain (SEQ ID NO: 85) MVSSAQFLGLLLLCFQGTRC DIVMTQTPSSLSASVGDRVTITC KASQDVI FDVA WYQQRPGQAPKLLIY SASSRVS GVPSRFSGSGSGTDFTLTISSLQP EDFATYYC QQHYSTPYT FGQGTRLDIK RTVAAPSVFIFPPSDEQLKSGTA SVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLT LSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
  • the changed residues in the humanized sequence are set out in Table 27 below (as VL variant 3, VH 3 , as well as the changed residues in three other humanized variants of the original 13E2 light chain sequence: VL variants 1, 2, and 4, VL 1 , VL 2 , and VL 4 ).
  • the light chain framework sequences of the humanized antibody are:
  • VL FR1 DIVMTQTPSSLSASVGDRVTITC
  • VL FR2 WYQQRPGQAPKLLIY
  • VL FR3 GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC
  • SEQ ID NO: 67 VL FR4: FGQGTRLDIK
  • CHO cells expressing LAG-3 on their surface were incubated with different concentrations of a chimeric 13E2-human antibody (referred to as Chim13E2IgG4) comprising the chimeric heavy and light chains described in Example 17 (heavy chain: 13E2IgG4mut; light chain: 13E2IgK), IMP761, or human IgG4 (as an isotype-matched negative control).
  • Chim13E2IgG4 chimeric 13E2-human antibody
  • Example 17 dasheavy chain: 13E2IgG4mut; light chain: 13E2IgK
  • IMP761 plasmin-associated antigen IgG4
  • human IgG4 as an isotype-matched negative control.
  • Secondary antibody goat anti-human IgG-FITC was used to detect the presence of antibodies on the surface of the CHO-LAG-3 + cells.
  • FITC mean fluorescence of intensity (MFI) was determined after analysis by flow cytometry.
  • a BiacoreTM surface plasmon resonance analysis was performed using the chimeric antibody Chim13E2IgG4 (comprising the chimeric heavy chain 13E2IgG4mut, and the chimeric light chain 13E2IgK, described in Example 17), or the humanized 13E2 antibody (IMP761) described in Example 18, covalently immobilized to a C1 sensor chip.
  • the coating was performed in 10 mM sodium acetate, pH 5.0, to reach 13 ⁇ 1 RU.
  • the recombinant human LAG-3Ig protein (IMP321) was then passed over the captured antibodies at 6 different concentrations, ranging from 0.078-2.5 nM, in analysis buffer (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.05% Tween 20) at 25° C., with regeneration in every cycle.
  • analysis buffer (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.05% Tween 20) at 25° C., with regeneration in every cycle.
  • the analysis was carried out on BiacoreTM T200 and the data was fitted using the kinetic global fit (Langmuir 1:1) model.
  • the kinetics parameters are recorded in Table 29, and represent the average of three runs.
  • CFSE-labelled PBMCs from healthy donors (0.2 ⁇ 10 6 cells/well in complete RPMI+10% FBS) were incubated with a pool of peptides covering the sequence of CMV pp35 in triplicate, with 300 ng/ml human IgG4 (isotype control), Chim13E2IgG4 or IMP761.
  • the T cell response was evaluated by measuring the proliferation, evaluated using a proliferation index (calculated as the sum of the percentage of CD8 + T cells under each division peak, multiplied by the division number), and the expression of CD25 at day 5 by flow cytometry.
  • the percentage inhibition for each antibody, compared with the isotype-matched negative control (huIgG4) was calculated based on the proliferation index values, or the percentage of CD25 + T cells within the CD8 + T cell population.
  • CFSE-labelled PBMCs from healthy donors (0.2 ⁇ 10 6 cells/well in complete RPMI+10% FBS) were incubated with a pool of peptides covering the sequence of CMV pp35 in triplicate, with different doses of Chim13E2IgG4, IMP761, or human IgG4 (an isotype-matched negative control).
  • the T cell response was evaluated by measuring the proliferation (CFSE dilution) at day 5 by flow cytometry. The percentage of CD8 + T cells for each division number was calculated for the different antibody doses used.
  • the inhibitory effect of each antibody increased as the dose increased from 10 ng/ml to 100 ng/ml antibody.
  • the inhibitory effect was similar to that of 100 ng/ml antibody.
  • the inhibitory effect of IMP761 was similar to that of Chim13E2IgG4 at all doses tested.
  • PBMC or NK cells are replaced with Jurkat cells stably expressing human Fc ⁇ RIIIa (the high-affinity V158 receptor) and NFAT-responsive element driving expression of a luciferase reporter gene.
  • Fc ⁇ RIIIa the high-affinity V158 receptor
  • NFAT-responsive element driving expression of a luciferase reporter gene.
  • a test antibody has ADCC activity, it will bind together a target cell and the Fc ⁇ RIIIa receptor of a Jurkat cell. Resulting activation of the signalling pathway downstream of the Fc ⁇ RIIIa receptor results in NFAT pathway activation thereby inducing luciferase reporter gene expression. Luciferase activity is quantified by luminescence read-out.
  • Anti-CD20 antibody and Raji cells, provided with the assay kit, were used as a positive control.
  • the anti-CD20 antibody was also tested on SEB-stimulated PBMCs. The assays were carried out, following the manufacturer's instructions, using 75,000 effector cells with 12,500 target cells.
  • Bio-Glo luciferase assay system was used, according to the manufacturer's instructions, to measure luminescence using a PerkinElmer EnVision 2103 luminometer (integration time of 0.5 sec/well).
  • results are shown in Table 33 below, and in FIG. 29 ( a ) .
  • the results are presented as the fold change in the relative luminescence unit (RLU), calculated by dividing the RLU obtained in the presence of the test antibody (at the maximal concentration recommended by the manufacturer, 3 ⁇ g/ml), by the RLU obtained without antibody.
  • RLU relative luminescence unit
  • the positive control anti-CD20 antibody showed significant ADCC activity against the Raji cells (a B cell line), and against SEB-stimulated PBMCs, which contain a small percentage of B cells.
  • This assay uses PBMCs stimulated for one day in X-Vivo 10 medium (Lonza) with 100 IU/ml of IL-2 (Roche) and CFSE-labelled PBMCs, stimulated with SEB for two days to allow the expression of LAG-3 on T cells.
  • the assay was carried out in X-Vivo 10 medium at a 50:1 effector:target ratio, with high dose (3 ⁇ g/ml) IMP761 or an isotype-matched negative control antibody, hIgG4. After 4 hours, the cell mixtures were harvested and stained for CD4, CD8, CD25 and LAG-3 using fluorochrome-conjugated antibodies.
  • results are shown in Table 34, and in FIGS. 29 ( b ) and ( c ) .
  • the results are presented as the percentage of living CD4 + or CD8 + cells in the PBMC population (b), and the percentage of living LAG-3 + CD4 + or LAG-3+CD8 + cells in the PBMC population (c).
  • IMP761 antibody does not reduce the percentage of CD8 + or CD4 + T cells in the PBMC population, or the percentage of LAG-3 + CD8 + cytotoxic T cells, or LAG-3 + CD4 + helper T cells in the PBMC population.
  • the isotype-matched negative control antibody, hIgG4 caused a slight reduction in the viability of T cells in the PBMC population, especially of activated T cells expressing LAG-3.
  • SEB-stimulated cells used as target cells, were incubated with 3 ⁇ g/ml of IMP761, an isotype-matched negative control antibody, hIgG4, a CDC-positive anti-CD3 control antibody (clone MEM-57, Cerdalane), or an isotype-matched negative control mouse antibody, mIgG2a, for 45 minutes in PBS, 0.5% BSA. Unbound antibodies were then washed away, and the cells were incubated with rabbit complement diluted by 3 volumes in RPMI medium for 1 hour at 37° C. The cells were stained for CD4, CD8, CD25 and LAG-3 using fluorochrome-conjugated antibodies. Cell viability in each blood cell population was then assessed by flow cytometry, after exclusion of cells labelled by 7-AAD.
  • results are shown in Table 35, and in FIG. 30 .
  • the results are presented as the percentage of living CD4 + or CD8 + in the PBMC population (a), and the percentage of living LAG-3 + CD4 + or LAG-3 + CD8 + cells in the PBMC population (b).
  • IMP761 antibody does not reduce the percentage of CD8 + or CD4 + T cells in the PBMC population, or the percentage of LAG-3 + CD8 + cytotoxic T cells, or LAG-3 + CD4 + helper T cells in the PBMC population.
  • the anti-CD3 positive control antibody did cause a decrease in the percentage of T cells in the PBMC population, and of activated T cells expressing LAG-3.
  • IMP761 antibody showed no cytotoxic activity in any of the short-term cytotoxicity assays described in (1)-(3) above.
  • the cytotoxicity of IMP761 against LAG-3-expressing T cells was also evaluated after culturing antigen-stimulated PBMCs for several days.
  • PBMCs from healthy donors 0.2 ⁇ 10 6 cells/well in complete RPMI+10% FBS
  • a pool of peptides covering the sequence of CMV pp35 in the presence of 300 ng/ml IMP761, or human IgG4 (as an isotype-matched negative control).
  • the percentage of CD8 + and CD4 + T cells gated in living lymphocytes, as well as the percentage of LAG-3 + cells in these T cell subsets was measured by flow cytometry.
  • IMP761 antibody does not reduce the percentage of CD8 + or CD4 + T cells in the lymphocyte population, or the percentage of LAG-3 + CD8 + cytotoxic T cells, or LAG-3 + CD4 + helper T cells in the lymphocyte population.
  • IMP761 antibody does not possess cytotoxic activity, so inhibition of antigen-induced T cell proliferation and activation by this antibody is not due to any cytotoxic activity against activated T cells.
US15/756,767 2015-09-02 2016-09-01 Anti-LAG-3 antibodies Active 2037-02-22 US11680104B2 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
GB1515572.4 2015-09-02
GBGB1515572.4A GB201515572D0 (en) 2015-09-02 2015-09-02 Anti-LAG-3 antibodies
GB1515572 2015-09-02
GBGB1612437.2A GB201612437D0 (en) 2016-07-18 2016-07-18 Anti-lag-3 antibodies
GB1612437.2 2016-07-18
GB1612437 2016-07-18
PCT/EP2016/070664 WO2017037203A1 (en) 2015-09-02 2016-09-01 Anti-LAG-3 Antibodies

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/070664 A-371-Of-International WO2017037203A1 (en) 2015-09-02 2016-09-01 Anti-LAG-3 Antibodies

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/312,629 Continuation US20230272107A1 (en) 2015-09-02 2023-05-05 Anti-lag-3 antibodies

Publications (2)

Publication Number Publication Date
US20190153112A1 US20190153112A1 (en) 2019-05-23
US11680104B2 true US11680104B2 (en) 2023-06-20

Family

ID=56852278

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/756,767 Active 2037-02-22 US11680104B2 (en) 2015-09-02 2016-09-01 Anti-LAG-3 antibodies
US18/312,629 Pending US20230272107A1 (en) 2015-09-02 2023-05-05 Anti-lag-3 antibodies

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/312,629 Pending US20230272107A1 (en) 2015-09-02 2023-05-05 Anti-lag-3 antibodies

Country Status (20)

Country Link
US (2) US11680104B2 (zh)
EP (2) EP3798234A1 (zh)
JP (3) JP7074341B2 (zh)
KR (1) KR20180042412A (zh)
CN (2) CN108699145A (zh)
AU (2) AU2016316730B2 (zh)
BR (1) BR112018004181A2 (zh)
CA (1) CA2995639A1 (zh)
DK (1) DK3344654T3 (zh)
ES (1) ES2842306T3 (zh)
HK (1) HK1256116A1 (zh)
IL (1) IL257798A (zh)
MX (2) MX2018002610A (zh)
PH (1) PH12018500369A1 (zh)
PL (1) PL3344654T3 (zh)
PT (1) PT3344654T (zh)
RU (2) RU2760582C2 (zh)
SG (1) SG10201913864RA (zh)
WO (1) WO2017037203A1 (zh)
ZA (1) ZA201801432B (zh)

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR072999A1 (es) 2008-08-11 2010-10-06 Medarex Inc Anticuerpos humanos que se unen al gen 3 de activacion linfocitaria (lag-3) y los usos de estos
ES2842306T3 (es) * 2015-09-02 2021-07-13 Immutep Sas Anticuerpos anti LAG-3
CA2992853A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific antibodies specific for pd1 and tim3
IL263834B2 (en) 2016-06-20 2024-01-01 Kymab Ltd Antibodies against PD-L1
KR102576042B1 (ko) 2016-10-11 2023-09-07 아게누스 인코포레이티드 항-lag-3 항체 및 이의 사용 방법
WO2018184964A1 (en) 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Immunoconjugates of an anti-pd-1 antibody with a mutant il-2 or with il-15
PE20191463A1 (es) 2017-04-05 2019-10-16 Hoffmann La Roche Anticuerpos biespecificos de union especifica pd1 y lag3
TWI707871B (zh) * 2017-04-05 2020-10-21 瑞士商赫孚孟拉羅股份公司 抗lag3抗體
CA3059468A1 (en) * 2017-04-27 2018-11-01 Tesaro, Inc. Antibody agents directed against lymphocyte activation gene-3 (lag-3) and uses thereof
BR112019022873A8 (pt) 2017-05-02 2023-04-11 Merck Sharp & Dohme Formulação, e, vaso ou dispositivo de injeção.
JOP20190260A1 (ar) 2017-05-02 2019-10-31 Merck Sharp & Dohme صيغ ثابتة لأجسام مضادة لمستقبل الموت المبرمج 1 (pd-1) وطرق استخدامها
WO2018222722A2 (en) 2017-05-30 2018-12-06 Bristol-Myers Squibb Company Compositions comprising an anti-lag-3 antibody or an anti-lag-3 antibody and an anti-pd-1 or anti-pd-l1 antibody
HRP20231457T1 (hr) 2017-05-30 2024-05-10 Bristol-Myers Squibb Company Liječenje lag-3-pozitivnih tumora
CN111050792A (zh) * 2017-08-30 2020-04-21 凡恩世制药公司 抗lag-3抗体及其用途
TW201927337A (zh) * 2017-12-22 2019-07-16 大陸商江蘇恆瑞醫藥股份有限公司 一種lag-3抗體醫藥組成物及其用途
WO2019129137A1 (zh) * 2017-12-27 2019-07-04 信达生物制药(苏州)有限公司 抗lag-3抗体及其用途
CN109970856B (zh) 2017-12-27 2022-08-23 信达生物制药(苏州)有限公司 抗lag-3抗体及其用途
WO2019179365A1 (en) * 2018-03-20 2019-09-26 WuXi Biologics Ireland Limited Novel anti-lag-3 antibody polypeptide
CN110343178B (zh) * 2018-04-03 2022-07-22 上海开拓者生物医药有限公司 抗人lag-3单克隆抗体及其应用
CN110606892B (zh) * 2018-06-14 2023-09-26 华博生物医药技术(上海)有限公司 一种高亲和力高生物活性的lag-3抗体及其应用
CN111620949A (zh) * 2019-02-28 2020-09-04 三生国健药业(上海)股份有限公司 结合人lag-3的抗体、其制备方法和用途
GB201906118D0 (en) 2019-05-01 2019-06-12 Immutep S A Anti-LAG-3 binding molecules
GB201906127D0 (en) 2019-05-01 2019-06-12 Immutep S A Assays
JP2022532490A (ja) 2019-05-13 2022-07-15 リジェネロン・ファーマシューティカルズ・インコーポレイテッド 癌の治療における有効性の増強のためのpd-1阻害剤とlag-3阻害剤の組み合わせ
US20220227887A1 (en) * 2019-06-11 2022-07-21 Ono Pharmaceutical Co., Ltd. Immunosuppressant
KR20220164548A (ko) * 2020-04-03 2022-12-13 소렌토 쎄라퓨틱스, 인코포레이티드 Lag3에 결합하는 조작된 항체
EP4157879A1 (en) 2020-05-27 2023-04-05 Agilent Technologies, Inc. Anti-human lag-3 antibodies and their use in immunohistochemistry (ihc)
EP4196502A1 (en) 2020-08-13 2023-06-21 Bristol-Myers Squibb Company Methods of redirecting of il-2 to target cells of interest

Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0054951B1 (en) 1980-12-24 1984-12-12 Chugai Seiyaku Kabushiki Kaisha Dibenz(b,f)(1,4)oxazepine derivatives, process for preparing the same, and pharmaceutical compositions comprising the same
WO1986001533A1 (en) 1984-09-03 1986-03-13 Celltech Limited Production of chimeric antibodies
US4703039A (en) 1984-04-10 1987-10-27 New England Deaconess Hospital Corporation Method of producing biologically active molecules having extended life time
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4866132A (en) 1986-04-17 1989-09-12 The Research Foundation Of State University Of New York Novel radiopaque barium polymer complexes, compositions of matter and articles prepared therefrom
EP0154316B1 (en) 1984-03-06 1989-09-13 Takeda Chemical Industries, Ltd. Chemically modified lymphokine and production thereof
US4873316A (en) 1987-06-23 1989-10-10 Biogen, Inc. Isolation of exogenous recombinant proteins from the milk of transgenic mammals
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
WO1991014438A1 (en) 1990-03-20 1991-10-03 The Trustees Of Columbia University In The City Of New York Chimeric antibodies with receptor binding ligands in place of their constant region
EP0519596A1 (en) 1991-05-17 1992-12-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
WO1993003769A1 (en) 1991-08-20 1993-03-04 THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES Adenovirus mediated transfer of genes to the gastrointestinal tract
WO1993006213A1 (en) 1991-09-23 1993-04-01 Medical Research Council Production of chimeric antibodies - a combinatorial approach
WO1993009239A1 (en) 1991-11-08 1993-05-13 Research Corporation Technologies, Inc. Adeno-associated virus-2 basal vectors
WO1993011161A1 (en) 1991-11-25 1993-06-10 Enzon, Inc. Multivalent antigen-binding proteins
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5256334A (en) 1988-09-08 1993-10-26 The Research Foundation Of The State University Of New York Homogeneous radiopaque polymer-organobismuth composites
WO1994012649A2 (en) 1992-12-03 1994-06-09 Genzyme Corporation Gene therapy for cystic fibrosis
EP0239400B1 (en) 1986-03-27 1994-08-03 Medical Research Council Recombinant antibodies and methods for their production
US5346981A (en) 1993-01-13 1994-09-13 Miles Inc. Radiopaque polyurethanes
WO1994028938A1 (en) 1993-06-07 1994-12-22 The Regents Of The University Of Michigan Adenovirus vectors for gene therapy sponsorship
WO1995000655A1 (en) 1993-06-24 1995-01-05 Mc Master University Adenovirus vectors for gene therapy
WO1995011984A2 (en) 1993-10-25 1995-05-04 Canji, Inc. Recombinant adenoviral vector and methods of use
US5476786A (en) 1987-05-21 1995-12-19 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
EP0401384B1 (en) 1988-12-22 1996-03-13 Kirin-Amgen, Inc. Chemically modified granulocyte colony stimulating factor
US5514548A (en) 1993-02-17 1996-05-07 Morphosys Gesellschaft Fur Proteinoptimerung Mbh Method for in vivo selection of ligand-binding proteins
WO1996016990A1 (en) 1994-12-02 1996-06-06 The Wellcome Foundation Limited Humanized antibodies to cd38
WO1996017951A2 (en) 1994-12-09 1996-06-13 Rpms Technology Limited Identification of genes responsible for in vivo survival of microorganisms
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
WO1996032478A1 (en) 1995-04-14 1996-10-17 Genentech, Inc. Altered polypeptides with increased half-life
WO1997003695A1 (en) 1995-07-21 1997-02-06 Applied Research Systems Ars Holding N.V. Methods for detecting, identifying, isolating, and selectively labelling and targeting th1 lymphocytes by means of the lag-3 protein
US5643575A (en) 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
WO1997035614A1 (en) 1996-03-27 1997-10-02 Crc For Biopharmaceutical Research Pty. Ltd. The use of antibodies against cd48 for the treatment of t and b cell lymphomas and leukemias
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
WO1997043316A1 (en) 1996-05-10 1997-11-20 Beth Israel Deaconess Medical Center, Inc. Physiologically active molecules with extended half-lives and methods of using same
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
US5747035A (en) 1995-04-14 1998-05-05 Genentech, Inc. Polypeptides with increased half-life for use in treating disorders involving the LFA-1 receptor
US5849860A (en) 1991-03-25 1998-12-15 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US5874304A (en) 1996-01-18 1999-02-23 University Of Florida Research Foundation, Inc. Humanized green fluorescent protein genes and methods
WO1999015553A2 (de) 1997-09-23 1999-04-01 Bundesrepublik Deutschland Letztvertreten Durch Den Direktor Des Robert-Koch-Instituts Ko-stimulierendes polypeptid von t-zellen, monoklonale antikörper sowie die herstellung und deren verwendung
US5919445A (en) 1996-10-09 1999-07-06 Council Of Agriculture Executive Yuan Use of green fluorescent protein to trace the infection of baculovirus in insects and to increase viral UV stability
US5939045A (en) 1994-12-22 1999-08-17 Nissan Chemical Industries, Ltd. Organic bismuth derivatives for X-ray imaging
WO1999043713A1 (en) 1998-02-25 1999-09-02 Lexigen Pharmaceuticals Corporation Enhancing the circulating half-life of antibody-based fusion proteins
US5958713A (en) 1995-01-31 1999-09-28 Novo Nordisk A/S Method of detecting biologically active substances by using green fluorescent protein
US5968738A (en) 1995-12-06 1999-10-19 The Board Of Trustees Of The Leland Stanford Junior University Two-reporter FACS analysis of mammalian cells using green fluorescent proteins
US5976877A (en) 1990-01-08 1999-11-02 Institut National De La Sante Et De La Recherche Medicale (Inserm) Proteins produced by human lymphocytes DNA sequence encoding these proteins and their pharmaceutical and biological uses
US5976796A (en) 1996-10-04 1999-11-02 Loma Linda University Construction and expression of renilla luciferase and green fluorescent protein fusion genes
US5985577A (en) 1998-10-14 1999-11-16 The Trustees Of Columbia University In The City Of New York Protein conjugates containing multimers of green fluorescent protein
WO1999058679A1 (en) 1998-05-09 1999-11-18 Glaxo Group Limited Antibodies to cd23, derivatives thereof, and their therapeutic uses
US6020192A (en) 1996-01-18 2000-02-01 University Of Florida Humanized green fluorescent protein genes and methods
WO2000009560A2 (en) 1998-08-17 2000-02-24 Abgenix, Inc. Generation of modified molecules with increased serum half-lives
US6046305A (en) 1997-12-12 2000-04-04 Macromed, Inc. Heterofunctionalized star-shaped poly(ethylene gycols) for protein modification
US6066476A (en) 1994-11-10 2000-05-23 The Regents Of The University Of California Modified green fluorescent proteins
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
US6165745A (en) 1992-04-24 2000-12-26 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
EP1158004A2 (en) 2000-05-18 2001-11-28 Japan Tobacco Inc. Human monoclonal antibody against a costimulatory signal transduction molecule AILIM and pharmaceutical use thereof
WO2002031240A2 (en) 2000-10-06 2002-04-18 Milliken & Company Face plate for spun-like textured yarn
EP1229125A1 (en) 1999-10-19 2002-08-07 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
WO2002060919A2 (en) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules with extended half-lives, compositions and uses thereof
US6447784B1 (en) 1997-09-10 2002-09-10 Vion Pharmaceuticals, Inc. Genetically modified tumor-targeted bacteria with reduced virulence
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
US20030153043A1 (en) 1997-05-21 2003-08-14 Biovation Limited Method for the production of non-immunogenic proteins
WO2003076567A2 (en) 2002-03-05 2003-09-18 Eli Lilly And Company Heterologous g-csf fusion proteins
EP1374902A1 (en) 2001-03-27 2004-01-02 Japan Tobacco Inc. Remedies for inflammatory bowel diseases
US20040131637A1 (en) 2001-03-09 2004-07-08 Chatfield Steven Neville Salmonella promoter for heterologous gene expression
WO2004078928A2 (en) 2003-02-28 2004-09-16 The Johns Hopkins University T cell regulation
WO2004081026A2 (en) 2003-06-30 2004-09-23 Domantis Limited Polypeptides
US20050053973A1 (en) 2001-04-26 2005-03-10 Avidia Research Institute Novel proteins with targeted binding
US20050089932A1 (en) 2001-04-26 2005-04-28 Avidia Research Institute Novel proteins with targeted binding
US20050164301A1 (en) 2003-10-24 2005-07-28 Avidia Research Institute LDL receptor class A and EGF domain monomers and multimers
WO2005077042A2 (en) 2004-02-09 2005-08-25 Human Genome Sciences, Inc. Albumin fusion proteins
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
WO2005103086A1 (en) 2004-04-23 2005-11-03 Bundesrepublik Deutschland letztvertreten durch das Robert-Koch-Institut vertreten durch seinen Präsidenten Method for the treatment of t cell mediated conditions by depletion of icos-positive cells in vivo
WO2006012508A2 (en) 2004-07-22 2006-02-02 Genentech, Inc. Method of treating sjögren's syndrome
WO2006014679A1 (en) 2004-07-21 2006-02-09 Glycofi, Inc. Immunoglobulins comprising predominantly a glcnac2man3glcnac2 glycoform
WO2007011041A1 (ja) 2005-07-22 2007-01-25 Kyowa Hakko Kogyo Co., Ltd. 遺伝子組換え抗体組成物
US7214775B2 (en) 1999-04-09 2007-05-08 Kyowa Hakko Kogyo Co., Ltd. Method of modulating the activity of functional immune molecules
US20070148165A1 (en) 2005-07-22 2007-06-28 Kyowa Hakko Kogyo Co., Ltd. Recombinant antibody composition
WO2008096158A2 (en) 2007-02-08 2008-08-14 Domantis Limited Antibody single variable domains against serum albumin
EP1987839A1 (en) 2007-04-30 2008-11-05 I.N.S.E.R.M. Institut National de la Sante et de la Recherche Medicale Cytotoxic anti-LAG-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
US7521051B2 (en) 2002-12-23 2009-04-21 Medimmune Limited Methods of upmodulating adaptive immune response using anti-PD-1 antibodies
US7632924B2 (en) 2004-06-18 2009-12-15 Ambrx, Inc. Antigen-binding polypeptides and their uses
WO2010019570A2 (en) 2008-08-11 2010-02-18 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
EP1075496B1 (en) 1998-05-08 2010-03-24 Cambridge Enterprise Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
US20110008331A1 (en) 2007-10-05 2011-01-13 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
US20110027295A1 (en) 2007-09-07 2011-02-03 The Johns Hopkins University Adenosine Receptor Agonists and Antagonists to Modulate T Cell Responses
WO2011014438A1 (en) 2009-07-31 2011-02-03 N.V. Organon Fully human antibodies to btla
WO2011016238A1 (en) 2009-08-06 2011-02-10 Immunas Pharma, Inc. Antibodies that specifically bind to a beta oligomers and use thereof
US20110070238A1 (en) 2007-04-30 2011-03-24 Immutep Parc Club Orsay Cytotoxic anti-lag-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
WO2011110621A1 (en) 2010-03-11 2011-09-15 Ucb Pharma, S.A. Biological products: humanised agonistic anti-pd-1 antibodies
WO2014008218A1 (en) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
US20140220021A1 (en) 2011-08-11 2014-08-07 Ono Pharmaceutical Co., Ltd. Therapeutic agent for autoimmune diseases comprising pd-1 agonist
WO2014140180A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property Development Limited Anti-lag-3 binding proteins
US8859739B2 (en) 2010-09-16 2014-10-14 Baliopharm Ag Anti-huTNFR1 antibody and methods of use thereof for treatment
WO2015138920A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
FR2688514A1 (fr) 1992-03-16 1993-09-17 Centre Nat Rech Scient Adenovirus recombinants defectifs exprimant des cytokines et medicaments antitumoraux les contenant.
ES2842306T3 (es) * 2015-09-02 2021-07-13 Immutep Sas Anticuerpos anti LAG-3

Patent Citations (120)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0054951B1 (en) 1980-12-24 1984-12-12 Chugai Seiyaku Kabushiki Kaisha Dibenz(b,f)(1,4)oxazepine derivatives, process for preparing the same, and pharmaceutical compositions comprising the same
US4816397A (en) 1983-03-25 1989-03-28 Celltech, Limited Multichain polypeptides or proteins and processes for their production
EP0120694B2 (en) 1983-03-25 2002-04-24 Celltech Therapeutics Limited Processes for the production of lg molecules
EP0125023B1 (en) 1983-04-08 1991-06-05 Genentech, Inc. Recombinant immunoglobulin preparations, methods for their preparation, dna sequences, expression vectors and recombinant host cells therefor
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
EP0154316B1 (en) 1984-03-06 1989-09-13 Takeda Chemical Industries, Ltd. Chemically modified lymphokine and production thereof
US4703039A (en) 1984-04-10 1987-10-27 New England Deaconess Hospital Corporation Method of producing biologically active molecules having extended life time
WO1986001533A1 (en) 1984-09-03 1986-03-13 Celltech Limited Production of chimeric antibodies
EP0194276B1 (en) 1984-09-03 1993-08-11 Celltech Therapeutics Limited Production of chimeric antibodies
EP0239400B1 (en) 1986-03-27 1994-08-03 Medical Research Council Recombinant antibodies and methods for their production
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4866132A (en) 1986-04-17 1989-09-12 The Research Foundation Of State University Of New York Novel radiopaque barium polymer complexes, compositions of matter and articles prepared therefrom
US5476786A (en) 1987-05-21 1995-12-19 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US4873316A (en) 1987-06-23 1989-10-10 Biogen, Inc. Isolation of exogenous recombinant proteins from the milk of transgenic mammals
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5256334A (en) 1988-09-08 1993-10-26 The Research Foundation Of The State University Of New York Homogeneous radiopaque polymer-organobismuth composites
EP0401384B1 (en) 1988-12-22 1996-03-13 Kirin-Amgen, Inc. Chemically modified granulocyte colony stimulating factor
US6180370B1 (en) 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US5976877A (en) 1990-01-08 1999-11-02 Institut National De La Sante Et De La Recherche Medicale (Inserm) Proteins produced by human lymphocytes DNA sequence encoding these proteins and their pharmaceutical and biological uses
WO1991014438A1 (en) 1990-03-20 1991-10-03 The Trustees Of Columbia University In The City Of New York Chimeric antibodies with receptor binding ligands in place of their constant region
US5849860A (en) 1991-03-25 1998-12-15 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
EP0519596A1 (en) 1991-05-17 1992-12-23 Merck & Co. Inc. A method for reducing the immunogenicity of antibody variable domains
WO1993003769A1 (en) 1991-08-20 1993-03-04 THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEALTH AND HUMAN SERVICES Adenovirus mediated transfer of genes to the gastrointestinal tract
WO1993006213A1 (en) 1991-09-23 1993-04-01 Medical Research Council Production of chimeric antibodies - a combinatorial approach
WO1993009239A1 (en) 1991-11-08 1993-05-13 Research Corporation Technologies, Inc. Adeno-associated virus-2 basal vectors
WO1993011161A1 (en) 1991-11-25 1993-06-10 Enzon, Inc. Multivalent antigen-binding proteins
US6350861B1 (en) 1992-03-09 2002-02-26 Protein Design Labs, Inc. Antibodies with increased binding affinity
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US6165745A (en) 1992-04-24 2000-12-26 Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
WO1994012649A2 (en) 1992-12-03 1994-06-09 Genzyme Corporation Gene therapy for cystic fibrosis
US5346981A (en) 1993-01-13 1994-09-13 Miles Inc. Radiopaque polyurethanes
US5514548A (en) 1993-02-17 1996-05-07 Morphosys Gesellschaft Fur Proteinoptimerung Mbh Method for in vivo selection of ligand-binding proteins
WO1994028938A1 (en) 1993-06-07 1994-12-22 The Regents Of The University Of Michigan Adenovirus vectors for gene therapy sponsorship
WO1995000655A1 (en) 1993-06-24 1995-01-05 Mc Master University Adenovirus vectors for gene therapy
WO1995011984A2 (en) 1993-10-25 1995-05-04 Canji, Inc. Recombinant adenoviral vector and methods of use
US5643575A (en) 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
US6066476A (en) 1994-11-10 2000-05-23 The Regents Of The University Of California Modified green fluorescent proteins
WO1996016990A1 (en) 1994-12-02 1996-06-06 The Wellcome Foundation Limited Humanized antibodies to cd38
WO1996017951A2 (en) 1994-12-09 1996-06-13 Rpms Technology Limited Identification of genes responsible for in vivo survival of microorganisms
US5939045A (en) 1994-12-22 1999-08-17 Nissan Chemical Industries, Ltd. Organic bismuth derivatives for X-ray imaging
US5958713A (en) 1995-01-31 1999-09-28 Novo Nordisk A/S Method of detecting biologically active substances by using green fluorescent protein
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
WO1996032478A1 (en) 1995-04-14 1996-10-17 Genentech, Inc. Altered polypeptides with increased half-life
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
US5747035A (en) 1995-04-14 1998-05-05 Genentech, Inc. Polypeptides with increased half-life for use in treating disorders involving the LFA-1 receptor
US6197524B1 (en) 1995-07-21 2001-03-06 Institute National De La Sante Et De La Recherche Medicale Methods for detecting, identifying, isolating, and selectively labelling and targeting TH1 lymphocyte by means of the LAG-3 protein
WO1997003695A1 (en) 1995-07-21 1997-02-06 Applied Research Systems Ars Holding N.V. Methods for detecting, identifying, isolating, and selectively labelling and targeting th1 lymphocytes by means of the lag-3 protein
US5968738A (en) 1995-12-06 1999-10-19 The Board Of Trustees Of The Leland Stanford Junior University Two-reporter FACS analysis of mammalian cells using green fluorescent proteins
US6020192A (en) 1996-01-18 2000-02-01 University Of Florida Humanized green fluorescent protein genes and methods
US5874304A (en) 1996-01-18 1999-02-23 University Of Florida Research Foundation, Inc. Humanized green fluorescent protein genes and methods
US5968750A (en) 1996-01-18 1999-10-19 The University Of Florida Research Foundation Inc. Humanized green fluorescent protein genes and methods
WO1997035614A1 (en) 1996-03-27 1997-10-02 Crc For Biopharmaceutical Research Pty. Ltd. The use of antibodies against cd48 for the treatment of t and b cell lymphomas and leukemias
WO1997043316A1 (en) 1996-05-10 1997-11-20 Beth Israel Deaconess Medical Center, Inc. Physiologically active molecules with extended half-lives and methods of using same
US5976796A (en) 1996-10-04 1999-11-02 Loma Linda University Construction and expression of renilla luciferase and green fluorescent protein fusion genes
US5919445A (en) 1996-10-09 1999-07-06 Council Of Agriculture Executive Yuan Use of green fluorescent protein to trace the infection of baculovirus in insects and to increase viral UV stability
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US20030153043A1 (en) 1997-05-21 2003-08-14 Biovation Limited Method for the production of non-immunogenic proteins
US6447784B1 (en) 1997-09-10 2002-09-10 Vion Pharmaceuticals, Inc. Genetically modified tumor-targeted bacteria with reduced virulence
WO1999015553A2 (de) 1997-09-23 1999-04-01 Bundesrepublik Deutschland Letztvertreten Durch Den Direktor Des Robert-Koch-Instituts Ko-stimulierendes polypeptid von t-zellen, monoklonale antikörper sowie die herstellung und deren verwendung
US6046305A (en) 1997-12-12 2000-04-04 Macromed, Inc. Heterofunctionalized star-shaped poly(ethylene gycols) for protein modification
WO1999043713A1 (en) 1998-02-25 1999-09-02 Lexigen Pharmaceuticals Corporation Enhancing the circulating half-life of antibody-based fusion proteins
EP1075496B1 (en) 1998-05-08 2010-03-24 Cambridge Enterprise Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
WO1999058679A1 (en) 1998-05-09 1999-11-18 Glaxo Group Limited Antibodies to cd23, derivatives thereof, and their therapeutic uses
WO2000009560A2 (en) 1998-08-17 2000-02-24 Abgenix, Inc. Generation of modified molecules with increased serum half-lives
US5985577A (en) 1998-10-14 1999-11-16 The Trustees Of Columbia University In The City Of New York Protein conjugates containing multimers of green fluorescent protein
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
US7214775B2 (en) 1999-04-09 2007-05-08 Kyowa Hakko Kogyo Co., Ltd. Method of modulating the activity of functional immune molecules
EP1229125A1 (en) 1999-10-19 2002-08-07 Kyowa Hakko Kogyo Co., Ltd. Process for producing polypeptide
EP1158004A2 (en) 2000-05-18 2001-11-28 Japan Tobacco Inc. Human monoclonal antibody against a costimulatory signal transduction molecule AILIM and pharmaceutical use thereof
WO2002031240A2 (en) 2000-10-06 2002-04-18 Milliken & Company Face plate for spun-like textured yarn
US6946292B2 (en) 2000-10-06 2005-09-20 Kyowa Hakko Kogyo Co., Ltd. Cells producing antibody compositions with increased antibody dependent cytotoxic activity
WO2002060919A2 (en) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules with extended half-lives, compositions and uses thereof
US20040131637A1 (en) 2001-03-09 2004-07-08 Chatfield Steven Neville Salmonella promoter for heterologous gene expression
EP1374902A1 (en) 2001-03-27 2004-01-02 Japan Tobacco Inc. Remedies for inflammatory bowel diseases
US20050053973A1 (en) 2001-04-26 2005-03-10 Avidia Research Institute Novel proteins with targeted binding
US20050089932A1 (en) 2001-04-26 2005-04-28 Avidia Research Institute Novel proteins with targeted binding
WO2003011878A2 (en) 2001-08-03 2003-02-13 Glycart Biotechnology Ag Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
WO2003076567A2 (en) 2002-03-05 2003-09-18 Eli Lilly And Company Heterologous g-csf fusion proteins
US7521051B2 (en) 2002-12-23 2009-04-21 Medimmune Limited Methods of upmodulating adaptive immune response using anti-PD-1 antibodies
US8551481B2 (en) * 2003-02-28 2013-10-08 The Johns Hopkins University Anti-cancer vaccine composition comprising an anti-CD223 antibody and kit comprising an anti-cancer vaccine and an anti-CD223 antibody
WO2004078928A2 (en) 2003-02-28 2004-09-16 The Johns Hopkins University T cell regulation
WO2004081026A2 (en) 2003-06-30 2004-09-23 Domantis Limited Polypeptides
US20050164301A1 (en) 2003-10-24 2005-07-28 Avidia Research Institute LDL receptor class A and EGF domain monomers and multimers
WO2005077042A2 (en) 2004-02-09 2005-08-25 Human Genome Sciences, Inc. Albumin fusion proteins
WO2005103086A1 (en) 2004-04-23 2005-11-03 Bundesrepublik Deutschland letztvertreten durch das Robert-Koch-Institut vertreten durch seinen Präsidenten Method for the treatment of t cell mediated conditions by depletion of icos-positive cells in vivo
US7632924B2 (en) 2004-06-18 2009-12-15 Ambrx, Inc. Antigen-binding polypeptides and their uses
WO2006014679A1 (en) 2004-07-21 2006-02-09 Glycofi, Inc. Immunoglobulins comprising predominantly a glcnac2man3glcnac2 glycoform
WO2006012508A2 (en) 2004-07-22 2006-02-02 Genentech, Inc. Method of treating sjögren's syndrome
US20070148165A1 (en) 2005-07-22 2007-06-28 Kyowa Hakko Kogyo Co., Ltd. Recombinant antibody composition
EP1921090A1 (en) 2005-07-22 2008-05-14 Kyowa Hakko Kogyo Co., Ltd. Genetically modified antibody composition
WO2007011041A1 (ja) 2005-07-22 2007-01-25 Kyowa Hakko Kogyo Co., Ltd. 遺伝子組換え抗体組成物
WO2008096158A2 (en) 2007-02-08 2008-08-14 Domantis Limited Antibody single variable domains against serum albumin
US20110070238A1 (en) 2007-04-30 2011-03-24 Immutep Parc Club Orsay Cytotoxic anti-lag-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
EP1987839A1 (en) 2007-04-30 2008-11-05 I.N.S.E.R.M. Institut National de la Sante et de la Recherche Medicale Cytotoxic anti-LAG-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
US20100233183A1 (en) 2007-04-30 2010-09-16 Frederic Triebel Cytotoxic anti-lag-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
WO2008132601A1 (en) 2007-04-30 2008-11-06 Immutep Cytotoxic anti-lag-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
US20160176965A1 (en) 2007-04-30 2016-06-23 Immutep Parc Club Orsay Cytotoxic anti-lag-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
US9244059B2 (en) 2007-04-30 2016-01-26 Immutep Parc Club Orsay Cytotoxic anti-LAG-3 monoclonal antibody and its use in the treatment or prevention of organ transplant rejection and autoimmune disease
US20110027295A1 (en) 2007-09-07 2011-02-03 The Johns Hopkins University Adenosine Receptor Agonists and Antagonists to Modulate T Cell Responses
US20110008331A1 (en) 2007-10-05 2011-01-13 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
WO2010019570A2 (en) 2008-08-11 2010-02-18 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
US20110150892A1 (en) 2008-08-11 2011-06-23 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3) and uses thereof
WO2011014438A1 (en) 2009-07-31 2011-02-03 N.V. Organon Fully human antibodies to btla
WO2011016238A1 (en) 2009-08-06 2011-02-10 Immunas Pharma, Inc. Antibodies that specifically bind to a beta oligomers and use thereof
US20110229461A1 (en) 2010-03-11 2011-09-22 Kerry Louise Tyson PD-1 Antibodies
WO2011110621A1 (en) 2010-03-11 2011-09-15 Ucb Pharma, S.A. Biological products: humanised agonistic anti-pd-1 antibodies
US8859739B2 (en) 2010-09-16 2014-10-14 Baliopharm Ag Anti-huTNFR1 antibody and methods of use thereof for treatment
US20140220021A1 (en) 2011-08-11 2014-08-07 Ono Pharmaceutical Co., Ltd. Therapeutic agent for autoimmune diseases comprising pd-1 agonist
WO2014008218A1 (en) 2012-07-02 2014-01-09 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
US20140093511A1 (en) 2012-07-02 2014-04-03 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2014140180A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property Development Limited Anti-lag-3 binding proteins
US20140286935A1 (en) 2013-03-15 2014-09-25 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
US20160017037A1 (en) 2013-03-15 2016-01-21 Glaxosmithkline Intellectual Property Development Limited Anti-lag-3 binding proteins
WO2015138920A1 (en) 2014-03-14 2015-09-17 Novartis Ag Antibody molecules to lag-3 and uses thereof
US9908936B2 (en) 2014-03-14 2018-03-06 Novartis Ag Antibody molecules to LAG-3 and uses thereof

Non-Patent Citations (221)

* Cited by examiner, † Cited by third party
Title
"Recombinant Antibodies for in vivo research use", http://absolutelyantibody.com/catalog/in-vivo-research, 2015.
"The European Committee on Antimicrobial Susceptibility Testing. Breakpoint tables for interpretation of MICs and zone diameters. Version 5.0, 2015. http://www.eucast.org."
Adelman, John P., et al."In Vitro Deletional Mutagenesis for Bacterial Production of the 20,000-Dalton Form of Human Pituitary Growth Hormone"; DNA; vol. 2, No. 3; 1983.
Adib-Conquy, Minou, et al."Effect of Amino Acid Substitutions in the Heavy Chain CDR3 of an Autoantibody on its Reactivity"; International Immunology; vol. 10, No. 3; pp. 341-346; Published 1998.
Albrecht, Huguette, et al."Monospecific Bivalent scFv-SH: Effects of Linker Length and Location of an Engineered Cysteine on Production, Antigen Binding Activity and Free SH Accessibility"; Journal of Immunologial Methods; 310; 2006; pp. 100-116.
Ali, Robin R., et al."Adeno-Associated Virus Gene Transfer to Mouse Retina"; Human Gene Therapy; 9:81-86; Jan. 1, 1998.
Alpuche-Aranda, Celia M., et al."Salmonella typhimurium Activates Virulence Gene Transcription within Acified Macrophage Phagosomes"; Proc Natl. Acad. Sci USA; vol. 89; pp. 10079-10083; Nov. 1992.
Altschul, Stephen F., et al."Gapped Blast and PSI-Blast: a New Generation of Protein Database Search Programs"; Nucleic Acids Research; vol. 25, No. 17; pp. 3389-3402; 1997.
Amit, A.G., et al."Three-Dimensional Structure of an Antigen-Antibody Complex at 2.8 Å Resolutions"; Science; vol. 233, pp. 747-753; Aug. 15, 1986.
Andre, et al., "CD40L stabilizes arterial thrombi by a B3 integrin-dependent mechanism," Nature Medicine, vol. 8, No. 3, pp. 247-252 (2002).
Andreae et al., "Maturation and Activation of Dendritic Cells Induced by Lymphocyte Activation Gene-3 (CD223)1", The Journal of Immunology, vol. 168, pp. 3874-3880 (2002).
Andreae, et al., "MHC class II signal transduction in human dendritic cells induced by a natural ligand, the LAG-3 protein (CD223)", Blood, vol. 102, No. 6, pp. 2130-2137 (2003).
Angal, S., et al."A Single Amino Acid Substitution Abolishes the Heterogeneity of Chimeric Mouse/Human (IgG4) Antibody"; Molecular Immunology; vol. 30, No. 1; pp. 105-108; Great Britain; 1993.
Annunziato, et al., "Expression and release of LAG-3-encoded protein by human CD4+ T cells are associated with IFN-γ production", The FASEB Journal, vol. 10, pp. 769-776 (1996).
Avice, et al., "Lymphocyte Activation Gene-3, a MHC Class II Ligand Expressed on Activated T Cells, Stimulates TNF-a and IL-12 Production by Monocytes and Dendritic CellsT", The Journal of Immunology, vol. 162, pp. 2748-2753 (1999).
Baixeras, Elena, et al."Characterization of the Lymphocyte Activation Gene 3-Encoded Protein. A New Ligand for Human Leukocyte Antigen Class II Antigens"; J. Exp Med.; vol. 176; pp. 327-337; Published Aug. 1, 1992.
Baixeras, et al., "Characterization of the Lymphocyte Activation Gene 3-Encoded Protein. A New Ligand for Human .leukocyte Antigen Class II Antigens", J. Exp. Med., vol. 176, pp. 327-337 (1992).
Bayry, et al., "Rescuing CD4+CD25+ regulatory T-cell functions in rheumatoid arthritis by cytokine-targeted monoclonal antibody therapy", Drug Discovery Today, vol. 12, No. 13/14, pp. 548-552 (2007).
Beers, Richard, et al."Immunotoxins with Increased Activity Against Epidermal Growth Factor Receptor vIII-expressing Cells Produced by Antibody Phage Display"; Clinical Cancer Research; vol. 6; pp. 2835-2843; Jul. 2000.
Belson, et al., "Characterisation of the clinical and activated T cell response to repeat delayed-type hypersensitivity skin challenges in human subjects, with KLH and PPD, as potential model to test T-cell targeted therapies", Inflammation Research, vol. 65, pp. 389-404 (2016).
Bennett, Jean, et al."Real-time, Noninvasive In Vivo Assessment of Adeno-Associated Virus-Mediated Retinal Transduction"; Investigative Opthalmology and Visual Science; vol. 38, No. 13; pp. 2857-2863; Dec. 1997.
Bettini, et al., "Cutting Edge: Accelerated Autoimmune Diabetes in the Absence of LAG-3", The Journal of Immunology, vol. 187, pp. 3493-3498 (2011).
Bindon, et al., "Importance of antigen specificity for complement-mediated lysis by monoclonal antibodies", Eur. J Immunol., vol. 18, pp. 1507-1514 (1988).
Bird, Robert E., et al."Single-Chain Antigen-Binding Proteins"; Science; vol. 242; pp. 423-426; Oct. 21, 1988.
Borras, T., et al."Adenoviral Reporter Gene Transfer to the Human Trabecular Meshwork Does Not Alter Aqueous Humor Outflow. Relevance for Potential Gene Therapy of Glaucoma"; Gene Therapy; vol. 6; pp. 515-524; 1999.
Bostrom, et al., "Chapter 19: Improving Antibody Binding Affinity And Specificity for Therapeutic Development", Therapeutic Antibodies: Methods and Protocols, vol. 525; pp. 353-376 (2009).
Boyd, et al., "The Effect of the Removal of Sialic Acid, Galactose and Total Carbohydrate on the Functional Activity of Campath-1H", Molecular Immunology, vol. 32, No. 17/18, pp. 1311-1318 (1995).
Brekke, Ole Henrik, et al."Therapeutic Antibodies for Human Diseases at the Dawn of the Twenty-First Century"; Nature Reviews Drug Discovery; pp. 52-62; vol. 2; Jan. 2003.
Brignone, et al., "A Soluble Form of Lymphocyte Activation Gene-3 (IMP321) Induces Activation of a Large Range of Human Effector Cytotoxic Cells", The Journal of Immunology, vol. 179, pp. 4202-4211 (2007).
Brummell, David A., et al."Probing the Combining Site of an Anti-Carbohydrate Antibody by Saturation-Mutagenesis: Role of the Heavy-Chain CDR3 Residues"; Biochemistry; vol. 32; pp. 1180-1187; Published 1993.
Bruniquel D. et al., "Regulation of Expression of the Human Lymphocyte Activation Gene-3 (LAG-3) Molecule, a LIgand for MHC Class II", Immunogenetics 48:116-124 (1998).
Buchholz, et al., "The smallest unit: effector and memory CD8+ T cell differentiation on the single cell level", Frontiers in Immunology, vol. 4, Article 31, pp. 1-10 (2013).
Buckner, Jane Hoyt, "Mechanisms of impaired regulation by CD4+CD25+FOXP3+ regulatory T cells in human autoimmune diseases", Nature Rev. Immunol., vol. 10, No. 12, pp. 849-859 (2010).
Camarero, Julio A., et al."Synthesis of Proteins by Native Chemical Ligation Using Fmoc-Based Chemistry"; Protein & Peptide Letters; vol. 12; pp. 723-728; 2005.
Camisaschi, et al., "LAG-3 Expression Defines a Subset of CD4+CD25highFoxp3+ Regulatory T Cells That Are Expanded at Tumor Sites", The Journal of Immunology, vol. 184, pp. 6545-6551 (2010).
Campbell, et al. "Collagen-induced arthritis in C57BL/6 (H-2b) mice: new insights into an important disease model of rheumatoid arthritis", Eur. J Immunol., vol. 30, pp. 1568-1575 (2000).
Caravan, Peter, et al."Gadolinium (III) Chelates as MRI Contrast Agents: Structure, Dynamics, and Applications"; Chem. Rev.; vol. 99; pp. 2293-2352; 1999.
Carrier, MJ, et al."Expression of Human IL-1 Beta in Salmonella typhimurium. A Model System for the Delivery of Recombinant Therapeutic Proteins in vivo"; The Journal of Immunology; vol. 148; pp. 1176-1181; 1992.
Chan, et al., "Variable region domain exchange in human IgGs promotes antibody complex formation with accompanying structural changes and altered effector functions", Molecular Immunology, vol. 41, pp. 527-538 (2004).
Chappel, et al., "Identification of a Secondary FcyRI Binding Site within a Genetically Engineered Human IgG Antibody", (1993) J Biological Chem 268(33); 25124-25131.
Chatfield, S.N., et al."Use of the nirB Promoter to Direct the Stable Expression of Heterologous Antigens in Salmonella Oral Vaccine Strains: Development of a Single-Dose Oral Tetanus Vaccine"; Bio/Technology; vol. 10; Aug. 1992.
Chavele and Ehrenstein, "Regulatory T-cells in systemic lupus erythematosus and rheumatoid arthritis", FEBS Letters, vol. 585, pp. 3603-3610 (2011).
Chen, Tracy "Formulation Concerns of Protein Drugs" Drug Development and Industrial Pharmacy: 18:11-12, 1311-1354; 1992.
Chothia, Cyrus, et al."Canonical Structures for the Hypervariable Regions of Immunoglobulins"; J. Mol. Biol.; vol. 196; pp. 901-917; 1987.
Chothia, Cyrus, et al."Conformations of Immunoglobulin Hypervariable Regions"; Nature; vol. 342, pp. 877-883; Dec. 1989.
Chothia, Cyrus, et al."Domain Association in Immunoglobulin Molecules. The Packing of Variable Domains"; J. Mol. Biol.: vol. 186; pp. 651-663; 1985.
Chothia, et al., "Conformations of immunoglobulin hypervariable regions", Nature, vol. 342, No. 21/28, pp. 877-883(1989).
Clinical Trial Summary, Safety, Tolerability, Efficacy and Dose-response of GSK2831781 in Ulcerative Colitis, https://clinicaltrials.gov.ct2/show/CCT03893565, 2019.
Co, MS, et al."Chimeric and Humanized Antibodies with Specificity for the CD33 Antigen"; The Journal of Immunology vol. 148; pp. 1149-1154; 1992.
Cole, et al., "The EBV-Hybridoma Technique and its Application to Human Lung Cancer", pp. 77-96, 1985.
Coles, et al., "Alemtuzumab vs. interferon Beta-1a in Early Multiple Sclerosis", The New England Journal of Medicine, vol. 359, No. 17, pp. 1786-1801 (2008).
Collin, "Immune checkpoint inhibitors: a patent review (2010-2015)", Expert Opinion on Therapeutic Patents, vol. 26, No. 5, pp. 555-564 (2016).
Cox, Jonathan P.L., et al."A Directory of Human Germ-Line Vx Segments Reveals a Strong Bias in their Usage"; Eur. J. Immunol. vol. 24; pp. 827-836; 1994.
Curtet, Chantal, et al."Polylysine-Gd-DTPAn and Polylysine-Gd-DOTAn Couple to Anti-CEA F(ab′)2 Fragments as Potential Immunocontrast Agents; Relaxometry, Biodistribution, and Magnetic Resonance Imaging in Nude Mice Grafted with Human Colorectal Carcinoma"; Investigative Radiology; vol. 33, Issue 10; pp. 752-761; Oct. 1998.
Dall' Acqua, et al., "Increasing the Affinity of a Human IgG1 for the Neonatal Fc Receptor: Biological Consequences", The Journal of Immunology, vol. 169, pp. 5171-5180 (2002).
Dall'Acqua, et al., "Modulation of the Effector Functions of a Human IgG1 through Engineering of its Hinge Region", The Journal of Immunology, vol. 177, pp. 1129-1138 (2006).
Daugherty, Bruce L., et al."Polymerase Chain Reaction Facilitates the Cloning, CDR-Grafting, and Rapid Expression of a Murine Monoclonal Antibody Directed Against the CD18 Component of Leukocyte Integrins"; Nucleic Acids Research; vol. 19, No. 9; pp. 2471-2476; 1991.
Davis, B.G., "Synthesis of Glycoproteins", Chem. Rev., vol. 102, pp. 579-601 (2002).
De Boer, Herman A., et al."The TAG Promoter: A Functional Hybrid Derived from the TRP and LAC Promoters"; Proc. Natl. Acad. Sci. USA; vol. 80; pp. 21-25; Jan. 1983.
De Groot and Martin, "Reducing Risk, Improving Outcomes: Bioengineering Less Immunogenic Protein Therapeutics", Clinical Immunology, vol. 131, pp. 189-201 (2009).
De Pascalis, et al. "Grafting of ‘Abbreviated’ Complementarity-Determining Regions Containing Specificity-Determining Residues Essential for Ligand Contact to Engineer a Less Immunogenic Humanized Monoclonal Antibody", The Journal of Immunology, vol. 169, pp. 3076-3084 (2002).
De Wildt, Ruud M.T., et al."Heavy Chain CDR3 Optimization of a Germline Encoded Recombinant Antibody Fragment Predisposed to Bind the U1A Protein"; Protein Engineering; vol. 10, No. 7; pp. 835-841; 1997.
Doria, et al., "Autoinflammation and autoimmunity: Bridging the divide", Autoimmunity Reviews, vol. 12, pp. 22-30 (2012).
Dotti, "The Other Face of Chimeric Antigen Receptors", Molecular Therapy, vol. 22, No. 5, pp. 899-900, (2014).
Drake C.G. et al., "Blocking the Regulatory T Cell Molecule LAG-3 Augments In Vivo Anti-Tumor Immunity in an Autochthonous Model of Prostate Cancer", Journal of Clinical Oncology 24(ISS):2573 (2006), abstract only.
Dunstan, Sarah J., et al."Use of In Vivo-Regulated Promoters to Deliver Antigens from Attenuated Salmonella enterica var. typhimurium"; Infection and Immunity; vol. 67, No. 10; pp. 5133-5141; Oct. 1999.
Edwards, B.M., et al. J. Mol. Biol. 2003;334:103-118 (Year: 2003). *
Ehrenstein, et al., "Compromised Function of Regulatory T Cells in Rheumatoid Arthritis and Reversal by Anti-TNFa Therapy", J. Exp. Med., vol. 200, No. 3, pp. 277-285 (2004).
Ellis et al. ‘Selective depletion of LAG3+ cells in T-cell-driven inflammation: a randomized, double-blind, placebo-controlled FTIH phase I/Ib clinical trial’ Poster Presentations, S336, 2018.
Ewert, et al., "Stability improvement of antibodies for extracellular and intracellular applications: CDR grafting to stable frameworks and structure-based framework engineering", Methods, vol. 34, pp. 184-199 (2004).
Flannery, John G., et al."Efficient Photoreceptor-Trageted Gene Expression in vivo by Recombinant Adeno-Associated Virus"; Proc. Natl. Acad. Sci. USA; vol. 94; pp. 6916-6921; Jun. 1997.
Flotte, Terence R., et al."Stable in vivo Expression of the Cystic Fibrosis Transmembrane Conductance Regulator with an Adeno-Associated Virus Vector"; Proc. Natl. Acad. Sci USA; vol. 90; pp. 10613-10617; Nov. 1993.
Freeman, et al., "Regulation of innate CD8+ T-cell activation mediated by cytokines", PNAS, vol. 109, No. 25, pp. 3971-9976, (2012).
Galli, et al., "Unequivocal Delayed Hypersensitivity in Mast Cell-Deficient and Beige Mice", Science, vol. 226, pp. 710-713 (1984).
Ganesan, A. "Solid-Phase Synthesis in the Twenty-First Century"; Mini-Reviews in Medical Chemistry; vol. 6 pp. 3-10; 2006.
Ghetie and Ward, "Multiple Roles for the Major Histocompatibility Complex Class I-Related Receptor FcRn", Annu. Rev. Immunol., vol. 18, pp. 739-766 (2000).
Ghetie, et al., "Increasing the serum persistence of an IgG fragment by random mutagenesis", Nature Biotechnol, vol. 15. pp. 637-640 (1997).
Hall, BL, et al."A Single Amino Acid Mutation in CDR3 of the 3-14-9 L Chain Abolished Expression of the IDA 10-defined Idiotope and Antigen Binding"; The Journal of Immunology; vol. 149; pp. 1605-1612; 1992.
Hang and Bertozzi, "Chemoselective Approaches to Glycoprotein Assembly", Acc. Chem. Res., vol. 34, pp. 727-736 (2001).
Hannier, et al., "CD3/TCR Complex-Associated Lymphocyte Activation Gene-3 Molecules Inhibit CD3/TCR Signaling", The Journal of Immunology, vol. 161, pp. 4058-4065 (1998).
Hannier, Sigrid, et al."CD3/TCR Complex-Associated Lymphocyte Activation Gene-3 Molecules Inhibit CD3/TCR Signaling"; The Journal of Immunology; vol. 161; pp. 4058-4065; 1998.
Harborne, Nerina R., et al."Transcriptional Control, Translation and Function of the Products of the Five Open Reading Frames of the Escherichia coli Nir Operon"; Moleculare Microbiology; vol. 6, No. 19; pp. 2805-2813; 1992.
Hargreaves, et al., "Selective depletion of activated T cells: the CD40L-specific antibody experience", Trends in Molecular Medicine, vol. 10, No. 3, pp. 130-135 (2004).
Haudebourg, et al., "Depletion of LAG-3 Positive Cells in Cardiac Allograft Reveals Their Role in Rejection and Tolerance", Transplantation, vol. 84, No. 11, pp. 1500-1506 (2007).
Herwijnen, et al., "Regulatory T cells that recognize a ubiquitous stress-inducible self-antigen are long-lived suppressors of autoimmune arthritis", PNAS, vol. 109, No. 35, pp. 14134-14139 (2012).
Hillen, Wolfgang, et al."Tet Repressor-tet Operator Interaction"; Protein Nucleic Acid Ineraction; vol. 10, pp. 143-162; 1989.
Hinton, et al., Engineered Human IgG Antibodies with Longer Serum Half-lives in Primates, J. Biological Chem., vol. 279, No. 8, pp. 6213-6216 (2004).
Hodgson, John, "Making Monoclonals in Microbes", Bio/Technology, vol. 9, pp. 421-425 (1991).
Holliger and Hudson, "Engineered Antibody Fragments and The Rise of Single Domains", Nature Biotechnology, vol. 23, No. 9, pp. 1126-1136 (2005).
Holliger, Philipp, et al."Diabodies: Small Bivalent and Bispecific Antibody Fragments"; Proc. Natl. Acad. Sci. USA; vol. 90; pp. 6444-6448; Jul. 1993.
Holt, Lucy J., et al."Domain Antibodies: Proteins for Therapy"; Trends in Biotechnology; vol. 21, No. 11 pp. 484-490; Nov. 2003.
Huang, et al., "Role of LAG-3 in Regulatory T Cells", Immunity, vol. 21, pp. 503-513 (2004).
Huard, Bertrand, et al."Characterization of the Major Histocompatibility Complex Class II Binding Site on LAG-3 Protein"; Proc. Natl. Acad. Sci. USA; vol. 94; pp. 5744-5749; May 1997.
Huard, Bertrand, et al."T Cell Major Histocompatibility Complex Class II Molecules Down-Regulate CD4+ T Cell Clone Responses Following LAG-3 Binding"; Eur. J. Immunol.; vol. 26; pp. 1180-1186; 1996.
Huard, et al., "Cellular expression and tissue distribution of the human LAG-3-encoded protein, an MHC class II Ligand", Immunogenetics, vol. 39, pp. 213-217 (1994).
Huard, et al., "Characterization of the major histocompatibility complex class II binding site on LAG-3 protein", Proc. Natl. Acad. Sci. USA, vol. 94, pp. 5744-5749 (1997).
Huard, et al., "LAG-3 does not define a specific mode of natural killing in human", Immunology Letters, vol. 61, pp. 109-112 (1998).
Huard, et al., "Lymphocyte-activation gene 3/major histocompatibility complex class II interaction modulates the antigenic response of CD4+ T lymphocytes", Eur J. Immunol, vol. 24, pp. 3216-3221 (1994).
Inglis, et al., "Collagen-Induced arthritis in C57BL/6 mice is associated with a robust and sustained T-cell response to type II collagen", Arthritis Research & Therapy, vol. 9, R113, pp. 1-8 (2007).
InvivoGen (www.invivogen.com)—Antibody Isotype Families (2015).
InvivoGen (www.invivogen.com)—Immunoglobulin G Review, 2011.
Iwai, et al., "Involvement of Inducible Costimulator-B7 Homologous Protein Costimulatory Pathway in Murine Lupus Nephritis I", J Immunol., vol. 171. pp. 2848-2854 (2003).
IYAD JAMALI, FIELD, ELIZABETH.H, FLEMING, AUDREY, COWDERY, JOHN.S: "KINETICS OF ANTI-CD4-INDUCED T HELPER CELL DEPLETION AND INHIBITION OF FUNCTION Activation of T Cells by the CD3 Pathway Inhibits Anti-CD4-Mediated T Cell Elimination and Down-Regulation of Cell Surface CD4", THE JOURNAL OF IMMUNOLOGY, AMERICAN ASSOCIATION OF IMMUNOLOGISTS, vol. 148, no. 6, 15 March 1992 (1992-03-15), pages 1613 - 1619, XP055213914, ISSN: 00221767
Jamali et al., "Kinetics of Anti-CD4-Induced T Helper Cell Depletion and Inhibition of Function. Activation of T Cells by the CD3 Pathway Inhibits Anti-CD4-Mediated T Cell Elimination and Down-Regulation of Cell Surface CD4", The Journal of Immunology, vol. 148, No. 6, Mar. 15, 1992 (Mar. 15, 1992), pp. 1613-1619, XP055213914, ISSN: 0022-1767.
Jomary, C., et al."Rescue of Photoreceptor Function by AAV-Mediated Gene Transfer in a Mouse Model of Inherited Retinal Degeneration"; Gene Therapy; vol. 4; pp. 683-690; 1997.
Jones, et al., Chapter 21; Deimmumization of Monoclonal Antibodies; Antony Dimitrov (Ed), Therapeutic Antibodies: Methods and Protocols 525; 405-423 (2009).
Jones, Peter T., et al."Replacing the Complementarity-determining Regions in a Human Antibody with Those from a Mouse"; Nature; vol. 321; May 1986; pp. 522-525.
Junghans, R.P., "Finally! The Brambell Receptor (FcRB), Mediator of Transmission of Immunity and Protection from Catabolism for IgG", Immunol Res 16(1): pp. 29-57 (1997).
Kabat, Elvin A., et al."Unusual Distribtions of Amino Acids in Complementarity-Determining (Hypervariable) Segments of Heavy and Light Chains of Immunoglobulins and Their Possible Roles in Specificity of Antibody-Combining Sites"; The Journal of Biological Chemistry; vol. 252, No. 19; pp. 6609-6616; Oct. 10, 1977.
Kammann, Matthias, et al."Rapid Insertional Mutagenesis of DNA by Polymerase Chain Reaction (PCR)"; Nucleic Acids Research; vol. 17, No. 13; 1989; p. 5404.
Kelley, Robert F., et al."Thermodynamic Analysis of an Antibody Functional Epitope"; Biochemistry; vol. 32; pp. 6828-6835; 1993.
Khan, T., and Salunke, D.M. J. Immunol. 2014;192:5398-5405 (Year: 2014). *
Kim, et al., "Antibody Engineering for the Development of Therapeutic Antibodies", Mol. Cells, vol. 20, No. 1, pp. 17-29 (2005).
Kisielow et al., "Expression of lymphocyte activation gene 3 (LAG-3) on B cells is induced by T cells", European Journal of Immunology, vol. 35, pp. 2081-2088 (2005).
Kisielow, et al, "Expression of lymphocyte activation gene 3 (LAG-3) on B cells is induced by T cells", Eur. J. Immunol., vol. 35, pp. 2081-2088 (2005).
Kitade, et al., "Early Presence of Regulatory Cells in Transplanted Rats Rendered Tolerant by Donor-Specific Blood Transfusion", J Immunol, 75:4963-4970 (2005).
Klechevsky, Eynav, et al."Cross-priming CD8+ T Cells by Targeting Antigens to Human Dendritic Cells Through DCIR"; Blood ; vol. 116, No. 10; Sep. 9, 2010; pp. 1685-1698.
Kohler et al., "Continuous cultures of fused cells secreting antibody of predefined specificity", Nature, vol. 256, pp. 495-497 (1975).
Komissarov, Andrey A., et al."Site-Specific Mutagenesis of a Recombinant Anti-Single-Stranded DNA Fab"; The Journal of Biological Chemistry; vol. 272, No. 43; pp. 26864-26870; Oct. 24, 1997.
Koretzky, "Multiple Roles of CD4 and CD8 in T Cell Activation", Journal of Immunology, vol. 185, pp. 2643-2644, (2010).
Kunstfeld, et al., "Induction of cutaneous delayed-type hypersensitivity reactions in VEGF-A transgenic mice results in chronic skin inflammation associated with persistent lymphatic hyperplasia", Blood, vol. 104, No. 4, pp. 1048-1057 (2004).
Laffleur, Brice, et al."Production of Human or Humanized Antibodies in Mice"; Antibody Method and Protocols, Methods in Molecular Biology; vol. 901; 2012; pp. 149-159.
Lamminmaki, et al., "Crystal Structure of a Recombinant Anti-estradiol Fab Fragment in Complex with 17B-Estradiol", The Journal of Biological Chemistry, vol. 276, No. 39, pp. 36687-36694 (2001).
Lange-Asschenfeldt, et al.,"Increased and prolonged inflammation and angiogenesis in delayed type hypersensitivity reactions elicited in the skin of thrombospondin-2-deficient mice", Blood, vol. 99, No. 2, pp. 538-545 (2002).
Lauffer, Randall B., et al."Preparation and Water Relaxation Properties of Proteins Labeled with Paramagnetic Metal Chelates"; Magnetic Resonance Imaging; vol. 3; pp. 11-16; 1985.
Lazaret et al., "Engineered antibody Fc variants with enhanced effector function", (2006) PNAS 103(11); 4005-4010.
Lefranc, Marie-Paule, et al."IMGT Unique Numbering for Immunoglobulin and T Cell Receptor Variable Domains and Ig Superfamily V-Like Domains"; Developmental and Comparative Immunology; vol. 27; p. 55-77; 2003.
Lefranc, Marie-Paule, et al."IMGT, the International ImMunoGeneTics Database"; Nucleic Acids Research; vol. 27, No. 1; pp. 209-212; 1999.
Legat, et al., "Vaccination with LAG-31g (IMP321) and Peptides Induces Specific CD4 and CD8 T-Cell Responses in Metastatic Melanoma Patients-Report of a Phase I/IIa Clinical Trial", Clinical Cancer Research, vol. 22, No. 6, 2016.
Leinhardt, et al., "Active tuberculosis in Africa is associated with reduced Th1 and increased Th2 activity in vivo", Eur J Immunol 32: 1605-1613 (2002).
Lewis, Alan P., et al."Immunoglobulin Complementarity-determining Region Grafting by Recombinant Polymerase Chain Reaction to Generate Humanised Monoclonal Antibodies"; Gene; vol. 101; pp. 297-302; 1991.
Li, Tiansen, et al."In Vivo Transfer of a Reporter Gene to the Retina Mediated by an Adenoviral Vector"; Investigative Ophthalmology & Visual Science; vol. 35, No. 5; Apr. 1994; pp. 2543-2549.
Li, Tiansen, et al."Phenotype Correction in Retinal Pigment Epithelium in Murine Mucopolysaccharidosis VII by Adenovirus-Mediated Gene Transfer"; Proc. Natl. Acad. Sci. USA; vol. 92; pp. 7700-7704; Aug. 1995.
Liu, et al., "Kaempferol attenuates imiquimod-induced psoriatic skin inflammation in a mouse model", British Society for Immunology, Clinical and Experimental Immunology, vol. 198, pp. 403-415 (2019).
MacCallum, et al., "Antibody-antigen interactions: Contact analysis and binding site topography", (1996) J Mol Biol. 262: 732-745.
MacCallum, Robert M., et al."Antibody-Antigen Interactions: Contact Analysis and Binding Site Topography"; J. Mol. Biol.; vol. 262; pp. 732-745; 1996.
Macon-Lemaitre, et al., "The negative regulatory function of the Lymphocyte-activation gene-3 co-receptor (CD223) on human T cells", Immunology, vol. 115, No. 2, pp. 170-178 (2005).
Macon-Lemaitre, Laetitia, et al."The Negative Regulatory Function of the Lymphocyte-Activiation Gene-3 Co-Receptor (CD223) on Human T Cells"; Immunology; vol. 115; pp. 170-178; 2005.
Mages, et al., "Molecular cloning and characterization of murine ICOS and identification of B7h as COS ligand", Eur. J Immunol, vol. 30, pp. 1040-1047 (2000).
Matz, Mikhail V., et al."Fluorescent Proteins from Nonbioluminescent Anthozoa Species"; Nature Biotechnology; vol. 17; Oct. 1999; pp. 969-973.
McKelvie, Nicola D., et al."Expression of Heterologous Antigens in Salmonella typhimurium Vaccine Vectors Using the in vivo-inducible, SPI-2 Promoter, ssaG"; Vaccine; vol. 22; pp. 3243-3255; 2004.
Melton, D.A., et al."Efficient in vitro Synthesis of Biologically Active RNA and RNA Hybridization Probes from Plasmids Containing a Bacteriophage SP6 Promoter"; Nucleic Acids Research; vol. 12, No. 18; 1984.
Mendelson, Ella, et al."Expression and Rescue of a Nonselected Marker from an Integrated AAV Vector";Virology; vol. 166; pp. 154-165; 1988.
Merrifield, R.B."Solid Phase Peptide Synthesis. I. The Synthesis of a Tetrapeptide"; J. Am. Chem. Soc.; vol. 85; pp. 2149-2156; Jul. 20, 1963.
Miyahira, Andrea, "Types of immune cells present in human PBMC", Sanguine Biosciences, 2012, http://technical.sanguinebio.com.
Miyoshi, Hiroyuki, et al."Stable and Efficient Gene Transfer into the Retina Using an HIV-Based Lentiviral Vector"; Proc. Natl. Acad. Sci. USA; vol. 94; pp. 10319-10323; Sep. 1997.
Monk, et al.,. "Fc-dependent depletion of activated T cells occurs through CD40L-specific antibody rather than costimulation blockade", Nature Medicine, vol. 9, No. 10, pp. 1275-1280 (2003).
Muller-Gartner, Hans W."lmaging Techniques in the Analysis of Brain Function and Behaviour"; Tibtech; vol. 16; Mar. 1998; pp. 122-130.
Murphy, Michael, et al."Using Biacore to Measure the Binding Kinetics of an Antibody-Antigen Interaction"; Current Protocols in Protein Science; Chapter 19, Unit 19.14; 2006.
Nechansky, et al., "Compensation of endogenous IgG mediated inhibition of antibody-dependent cellular cytotoxicity by glyco-engineering of therapeutic antibodies", (2007) Molecular Immunol., vol. 44; 1815-1817.
Nicholson, et al., "Monocyte Dependent Regulation of Autoimmune Inflammation", Current Molecular Medicine, vol. 9, pp. 23-29, 2009.
Nishida, et al., "Novel humanized anti-CD20 monoclonal antibodies with unique germline VH and VL gene recruitment and potent effector functions", International Journal of Oncology, vol. 32, pp. 1263-1274 (2008).
Nobel-Jamieson, et al., "Auto-immune cholangiopathy in a juvenile patient with systemic lupus erythematosus", Acta Paediatrica, Foundation Acta Paediatrica, 10 1.e262-264 (2012).
Nobel-Jamieson, et al., "Auto-immune cholangiopathy in a juvenile patient with systemic lupus erythematosus", Acta Paediatrica, Foundation Acta Paediatrica, 101, pp. e262-e264 (2012).
Ono, et al., "Improved technique of heart transplantation in rats", Journal of Thoracic and Cardiovascular Surgery, vol. 57, No. 2, pp. 225-229 (1969).
Ozkaynak, et al., "Importance of iCOS-B7RP-1 costimulation in acute and chronic allograft rejection", Nature Immunology, vol. 2, No. 7, pp. 591-596 (2001).
Patel and Boyd, "An improved assay for antibody dependent cellular cytotoxicity based on time resolved fluorometry", (1995) J Immunol Meth 184:29-38.
Peggs, et al., "Cancer immunotherapy: co-stimulatory agonists and co-inhibitory antgaonists", The Journal of Translational Immunology, vol. 157, pp. 9-19 (2009).
Pluckthein, A., et al."The Pharmacology of Monoclonal Antibodies"; vol. 113; New York; pp. 269-315; 1994.
Pluckthun, Andreas, "Mono-and Bivalent Antibody Fragments Produced in Escherichia coli Engineering, Folding and Antigen Binding", (1992) Immunol Rev, 130; 151-188.
Poirier et al., "Antibody-mediated depletion of lymphocyte-activation gene-3 (LAG-3+)-activated T lymphocytes Prevents delayed-type hypersensitivity in non-human primates", Clin Experimental Immunol., 164(2): 265-274 (2011).
Poosarla, V.G., et al. Biotechnol. Bioengin. 2017; 114(6): 1331-1342 (Year: 2017). *
Pulkkinen, Wendy S., et al."A Salmonella typhimurium Virulence Protein is Similar to a Yersinia Enterocolitica Invasion Protein and a Bacteriophage Lambda Outer Membrane Protein"; Journal of Bacteriology; vol. 173, No. 1; pp. 86-93; Jan. 1991.
Queen, Cary, et al."A Humanized Antibody that Binds to the Interleukin 2 Receptor"; Proc. Natl. Acad. Sci. USA; vol. 86; pp. 10029-10033; Dec. 1989.
Queen, Cary, et al."Cell-Type Specific Regulation of a K Immunoglobulin Gene by Promoter and Enhancer Elements"; Immunological Reviews; Copenhagen, Denmark; No. 89; 1986; pp. 49-53.
Queen, et al., "A humanized antibody that binds to the interleukin 2 receptor", (1989) Proc Natl Acad Sci USA, 86: 10029-10032.
Raju, et al., "Glycoengineering of Therapeutic Glycoproteins: In Vitro Galactosylation and Sialylation of Glycoproteins with Terminal N-Acetylglucosamine and Galactose Residues", (2001) Biochem 40; 8868-8876.
Reddy, Manjula P., et al."Elimination of Fc Receptor-Dependent Effector Functions of a Modified IgG4 Monoclonal Antibody to Human CD4"; The Journal of Immunology; vol. 164; pp. 1925-1933; 2000.
Riechmann, Lutz, et al."Reshaping Human Antibodies for Therapy"; Nature; vol. 332;pp. 323-327; Mar. 24, 1988.
Rolling, Fabienne, et al."Evalution of Adeno-Associated Virus-Mediated Gene Transfer into the Rat Retina by Clinical Fluorescence Photography"; Human Gene Therapy; vol. 10; pp. 641-648; Mar. 1, 1999.
Rothe, et al., "Ribosome display for improved biotherapeutic molecules", (2006) Expert Opinion on Biological Therapy 6(2); 177-187.
Rudikoff S. et al., "Single Amino Acid Substitution Altering Antigen-Binding Specificity", Pro. Natl. Acad. Sci. USA 79:1979-1983 (Mar. 1982).
Saito, et al., "A Tumor Necrosis Factor Receptor Loop Peptide Mimic Inhibits Bone Destruction to the Same Extent as Anti-Tumor Necrosis Factor Monoclonal Antibody in Murine Collagen-Induced Arthritis", Arthritis & Rheumatism, vol. 56, No. 4, pp. 1164-1174 (2007).
Saji, Hideo "Targeted Delivery of Radiolabeled Imaging and Therapeutic Agents: Bifunctional Radiopharmaceuticals"; Critical Reviews in Therapeutic Drug Carrier Systems; vol. 16, No. 2; pp. 209-244; 1999.
Sakamoto, et al., "AILIM/ICOS: Its Expression and Functional Analysis with Monoclonal Antibodies", Hybridoma and Hybridomics, vol. 20, No. 5, pp. 293-303 (2001).
Saldanha, Jose W., Molecular Engineering I; Humanization—found in Chapter 6, pp. 119-144 of The Handbook of Therapeutic Antibodies, edited by Stephan Dubel, Jan. 1, 2007, Wiley-VCH, Weinheim, XP007913671, ISBN; 978-3-527-31453-9.
Samulski, Richard Jude, et al."Helper-Free Stocks of Recombinant Adeno-Associated Viruses: Normal Integration Does Not Require Viral Gene Expression"; Journal of Virology; vol. 63, No. 9; pp. 3822-3828; Sep. 1989.
Sato, Koh, et al."Reshaping a Human Antibody to Inhibit the Interleukin 6-dependent Tumor Cell Growth"; Cancer Research; vol. 53; pp. 851-856; Feb. 15, 1993.
Sears and Wong, "Toward Automated Synthesis of Oligosaccharides and Glycoproteins", (2001) Science 291; 2344-2350.
Seok, et al., "Genomic responses in mouse models poorly mimic human inflammatory diseases", PNAS, vol. 110, No. 9, pp. 3507-3512, Feb. 26, 2013.
Shetron-Rama, Lynne M., et al."Intracellular Induction of Listeria Monocytogenes actA Expression"; Infection and Immunity; vol. 70, No. 3; pp. 1087-1096; Mar. 2002.
Shields, et al., "High Resolution Mapping of the Binding Site on Human IgG1 for FcyRI, FcyRII, FcyRIII, and FcRn and Design of IgG1 Variants with Improved Binding to the FcyR", (2001) J Biol Chem 276(9); 6591-6604.
Siawaya, et al., "Immune parameters as markers of tuberculosis extent of disease and early prediction of anti-tuberculosis chemotherapy response", (2008) J Infect 56: 340-347.
Sierro, et al., "The CD4-like molecule LAG-3, biology and therapeutic applications" Expert Opinion on Therapeutic Targets 15(1): 91-101 (2011).
Sipkins, Dorothy A., et al."Detection of Tumor Angiogenesis in vivo by avB3-targeted Magnetic Resonance Imaging"; Nature Medicine; vol. 4, No. 5; May 1998; pp. 623-626.
Sizemore, Donata R., et al."Attenuated Shigella as a DNA Delivery Vehicle for DNA-Mediated Immunization"; Science; vol. 270; Oct. 13, 1995; pp. 299-302.
Slevin et al. ‘Lymphocytle Activation Gene (LAG)-3 is Associated with Mucosal Inflammation and Disease Activity in Ulcerative Colitis’ Journal of Crohn's and Colitis, Jan. 16, 2020.
Song, et al., "Rat and Human Natural Killers Exhibit Contrasting Immunoglobulin G Subclass Specificities in Antibody-Dependent Cellular Cytotoxicity Reflecting Differences in Their Fc Receptors FcyR)", Journal of Leukocyte Biology, vol. 48, pp. 524-530 (1990).
Sporici, et al., "ICOS Ligand Costimulation is Required for T-Cell Encephalitogenicity", Clinical Immunology, vol. 100, No. 3, pp. 277-288 (2001).
Ssacs, J D, "From bench to bedside; discovering rules for antibody design, and improving serotherapy with monoclonal antibodies", Rheumatology, vol. 40, pp. 724-738 (2001).
Stasiuk, et al., "Collagen-Induced Arthritis in DBA/1 Mice: Cytokine Gene Activation Following Immunization with Type II Collagen", Cellular Immunology, vol. 173, pp. 269-275 (1996).
STEFAN DÜBEL: "Handbook of Therapeutic Antibodies", 1 January 2007, WILEY-VCH , Weinheim , ISBN: 978-3-527-31453-9, article STEPHAN DUEBEL: "Handbook of Therapeutic Antibodies Chapter 6", pages: 119 - 144, XP007913671
Steplewski et al., "Biological activity of human-mouse IgG1, IgG2, IgG3, and IgG4 chimeric monoclonal antibodies with antitumor specificity", (1988) Proc Nat'l Acad Sci 85:4852-4856.
Stewart, et al., "The role of Fc gamma receptors in the activity of immunomodulatory antibodies for cancer", Journal for ImmunoTherapy of Cancer, vol. 2, No. 29, (2014).
Takahashi, Masayo, et al."Rescue from Photoreceptor Degeneration in the rd Mouse by Human Immunodeficiency Virus Vector-Mediated Gene Transfer"; Journal of Virology; vol. 73, No. 9; pp. 7812-7816; Sep. 1999.
T-Cell Research, T Cell Brochure, 2014, pp. 1-14, BD Biosciences, https://www.bdbiosciences.com > documents > cell.
Teeling, et al., "The Biological Activity of Human CD20 Monoclonal Antibodies is Linked to Unique Epitopes on CD201", The Journal of Immunology, vol. 177, pp. 362-371 (2006).
Thie, et al., Chapter 16: "Affinity Maturation by Phage Display", Antony Dimitrov (Ed). Therapeutic Antibodies; Methods and Protocols 525; 309-322 (2009) [Methods Mol Biol (2011).
Thomson, et al., "FK506; a novel immunosuppressant for treatment of autoimmune disease; Rationale and preliminary clinical experience at the University of Pittsburgh", Springer Semin Immunopathol. 14(4); 323-344 (1993).
Tomlinson I.M. et al., "The Repertoire of Human Germline VH Sequences Reveals About Fifty Groups of VH Segments With Different Hypervariable Loops", J. Mol. Biol. 227:776-798 (1992).
Torres, M., and Casadevall, A. Trends Immunol. 2008;91 -97 (Year: 2008). *
Totsuka, et al., "Ameliorating Effect of Anti-inducible Costimulator Monoclonal Antibody in a Murine Model of Chronic Colitis", Gastroenterology, vol. 124, pp. 410-421 (2003).
Townsend S. et al., "Augmented Binary Substitution: Single-Pass CDR Germ-Lining and Stabilization of Therapeutic Antibodies", PNAS 112(50):15354-15359 (Dec. 15, 2015).
Triebel, "LAG-3: a regulator of T-cell and DC responses and its use in therapeutic vaccination", Trends in Immunology, vol. 24, No. 12, pp. 619-622 (2003).
Triebel, et al., "A soluble lymphocyte activation gene-3 (sLAG-3) protein as a prognostic factor in human breast cancer expressing estrogen or progesterone receptors", (2006) Cancer Letters 235; 147-153.
Triebel, Frederic "LAG-3: A Regulator of T-cell and DC Responses and its Use in Therapeutic Vaccination"; Trends in Immunology; vol. 24, No. 12; Dec. 2003; pp. 619-622.
Uchida et al., "The innate mononuclear phagocyte network depletes B lymphocytes through Fc receptor-dependent mechanisms during anti-CD20 antibody immunotherapy", The Journal of Experimental Medicine, vol. 199, No. 12, pp. 1659-1669 (2004).
Valdivia, Raphael H., et al."Bacterial Genetics by Flow Cytometry; Rapid Isolation of Salmonella typhimurium Acid-Inducible Promoters by Differential Fluorescence Induction"; Molecular Microbiology; vol. 22, No. 2; pp. 367-378; 1996.
Velders et al., "The Impact of Antigen Density and Antibody Affinity on Antibody-Dependent Cellular Cytotoxicity: Relevance for Immunotherapy of Carcinomas", British Journal of Cancer, vol. 78, No. 4, pp. 478-483 (1998).
Virella, et al., "Biosynthesis, Metabolism, and Biological Properties", Introduction to Medical Immunology, Chapter 6, 4th Edition, pp. 99-100 (1998).
Wacker et al., "N-Linked Glycosylation in Campylobacter jejuni and its Functional Transfer into E. coli", Science, 298; 1790-1793 (2002).
Waldmann, Herman, "The new immunosuppression: just kill the T cell", Nature Medicine, vol. 9, No. 10, pp. 1259-1260 (2003).
Woo, et al., "Differential subcellular localization of the regulatory T-cell protein LAG-3 and the coreceptor CD4", Eur J Immunol, 40;1768-1777 (2010).
Workman and Vignali, "The CD4-related molecule, LAG-3 (CD223), regulates the expansion of activated T cells", Eur J Immunol, 33; 970-979 (2003).
Worn and Pluckthun, "Stability Engineering of Antibody Single-chain Fv Fragments", J Mol Biol, 305; 989-1010 (2001).
Yamane-Ohnuki, et al., "Establishment of FUT8 knockout Chinese hamster ovary cells: an ideal host cell line for producing completely defucosylated antibodies with enhanced antibody-dependent cellular cytotoxity", Biotechnology and Bioengineering 87(5); 614-622 (2004).
Yasuda, et al. "Humanization of Murine Antibodies by CDR-Grafting", Japanese Journal of Thrombosis and Hemostasis, 4(3): 193-200 (1993). (Translation attached).
Yousaf, et al., "Targeting behavior of rat monoclonal IgG antibodies in vivo: role of antibody isotype, specificity and the target cell antigen density", Eur. J Immunol, vol. 21, pp. 943-950 (1991).
Zapata, Gerardo, et al."Engineering Linear F(ab′)2 Fragments for Efficient Production in Escherichia coli and Enhanced Antiproliferative Activity"; Protein Engineering; vol. 8, No. 10; pp. 1057-1062; 1995.
Zhang, et al., "A New Strategy for the Synthesis of Glycoproteins", Science, 303; 371-373 (2004).
Zhang, et al., "CD8+ T Cells: Foot Soldiers of the Immune System", Immunity Review, vol. 35, pp. 161-168 (2011).

Also Published As

Publication number Publication date
AU2023204589A1 (en) 2023-08-03
EP3344654A1 (en) 2018-07-11
WO2017037203A1 (en) 2017-03-09
SG10201913864RA (en) 2020-03-30
PT3344654T (pt) 2021-01-26
RU2021133819A (ru) 2021-12-10
RU2018111508A3 (zh) 2020-02-18
CN108699145A (zh) 2018-10-23
JP7074341B2 (ja) 2022-05-24
JP2022137218A (ja) 2022-09-21
PL3344654T3 (pl) 2021-05-17
CA2995639A1 (en) 2017-03-09
RU2018111508A (ru) 2019-10-04
MX2018002610A (es) 2018-09-27
ES2842306T3 (es) 2021-07-13
RU2760582C2 (ru) 2021-11-29
JP2021097706A (ja) 2021-07-01
KR20180042412A (ko) 2018-04-25
EP3798234A1 (en) 2021-03-31
US20190153112A1 (en) 2019-05-23
PH12018500369A1 (en) 2018-09-03
CN117510633A (zh) 2024-02-06
ZA201801432B (en) 2019-01-30
BR112018004181A2 (pt) 2018-09-25
AU2016316730B2 (en) 2023-05-25
AU2016316730A1 (en) 2018-03-22
HK1256116A1 (zh) 2019-09-13
DK3344654T3 (da) 2021-01-18
US20230272107A1 (en) 2023-08-31
EP3344654B1 (en) 2020-10-21
JP2018537400A (ja) 2018-12-20
MX2023001226A (es) 2023-03-03
IL257798A (en) 2018-06-28

Similar Documents

Publication Publication Date Title
US20230272107A1 (en) Anti-lag-3 antibodies
EP2914291B1 (en) Anti-complement c1s antibodies and uses thereof
US11851482B2 (en) Anti-complement factor Bb antibodies and uses thereof
US20230265187A1 (en) Anti-tigit antibody and methods of use thereof
US20220306762A1 (en) Anti-cd38 antibody and methods of use thereof
CA3142634A1 (en) Anti-b7-h3 antibody and methods of use thereof

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: IMMUTEP S.A.S, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TRIEBEL, FREDERIC;BRIGNONE, CHRYSTELLE;REEL/FRAME:048341/0881

Effective date: 20180619

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO MICRO (ORIGINAL EVENT CODE: MICR); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: SMAL); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FEPP Fee payment procedure

Free format text: PETITION RELATED TO MAINTENANCE FEES GRANTED (ORIGINAL EVENT CODE: PTGR); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCF Information on status: patent grant

Free format text: PATENTED CASE

AS Assignment

Owner name: IMMUTEP S.A.S., FRANCE

Free format text: CHANGE OF ADDRESS;ASSIGNOR:IMMUTEP S.A.S.;REEL/FRAME:063906/0873

Effective date: 20221101